EP2619574A2 - Test de diagnostic moléculaire du cancer - Google Patents

Test de diagnostic moléculaire du cancer

Info

Publication number
EP2619574A2
EP2619574A2 EP11825959.7A EP11825959A EP2619574A2 EP 2619574 A2 EP2619574 A2 EP 2619574A2 EP 11825959 A EP11825959 A EP 11825959A EP 2619574 A2 EP2619574 A2 EP 2619574A2
Authority
EP
European Patent Office
Prior art keywords
cancer
dna
biomarkers
gene
expression
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP11825959.7A
Other languages
German (de)
English (en)
Other versions
EP2619574B1 (fr
EP2619574A4 (fr
Inventor
Timothy Davison
Jude O'donnell
Max Bylesjo
Fionnuala Mcdyer
Steve Deharo
Laura A. Hill
Katherine E. Keating
Vitali Proutski
Denis Paul Harkin
Richard Kennedy
Nicolas Goffard
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Almac Diagnostic Services Ltd
Original Assignee
Almac Diagnostics Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Almac Diagnostics Ltd filed Critical Almac Diagnostics Ltd
Priority to EP15181181.7A priority Critical patent/EP2975399B1/fr
Publication of EP2619574A2 publication Critical patent/EP2619574A2/fr
Publication of EP2619574A4 publication Critical patent/EP2619574A4/fr
Application granted granted Critical
Publication of EP2619574B1 publication Critical patent/EP2619574B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57449Specifically defined cancers of ovaries
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/582Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with fluorescent label
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • G16B25/10Gene or protein expression profiling; Expression-ratio estimation or normalisation
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B40/00ICT specially adapted for biostatistics; ICT specially adapted for bioinformatics-related machine learning or data mining, e.g. knowledge discovery or pattern finding
    • G16B40/20Supervised data analysis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/30ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for calculating health indices; for individual health risk assessment
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/04Methods of screening libraries by measuring the ability to specifically bind a target molecule, e.g. antibody-antigen binding, receptor-ligand binding

Definitions

  • the present invention relates to a molecular diagnostic test useful for diagnosing cancers from different anatomical sites that includes the use of a common DNA damage repair deficiency subtype.
  • the invention includes the use of a 44-gene classification model that is used to identify this DNA damage repair deficiency molecular subtype.
  • One application is the stratification of response to, and selection of patients for breast cancer therapeutic drug classes, including DNA damage causing agents and DNA repair targeted therapies.
  • Another application is the stratification of ovarian cancer patients into those that respond and those that do not respond to DNA damage causing agents.
  • the present invention provides a test that can guide conventional therapy selection as well as selecting patient groups for enrichment strategies during clinical trial evaluation of novel therapeutics.
  • DNA repair deficient subtypes can be identified from fresh/frozen (FF) or formalin fixed paraffin embedded (FFPE) patient samples.
  • Standard chemotherapy typically includes direct DNA damaging agents such as anthracyclines and alkylating agents as well as antimetabolites and antimicrotubule agents.
  • Ovarian cancer is the leading cause of death among all gynecological cancers in western countries. This high death rate is due to the diagnosis at an advanced stage in most patients.
  • Epithelial ovarian cancer (EOC) constitutes 90% of ovarian malignancies and is classified into distinct histologic categories including serous, mucinous, endometrioid, clear cell, transitional, mixed, and undifferentiated subtypes. There is increasing evidence that these differed histologies arise from different aetiologies.
  • the current standard treatment for ovarian cancer is debulking surgery and standard platinum taxane based cytotoxic chemotherapy. However, not all patients respond to this, and of those that do, approximately 70% will experience a recurrence.
  • Specific targeted therapies for ovarian cancer based on histological or molecular classification have not yet reached the marketplace. Similarly for other types of cancer, there is still no accurate way of selecting appropriate cytotoxic chemotherapeutic agents.
  • microarrays and molecular genomics have the potential for a significant impact on the diagnostic capability and prognostic classification of disease, which may aid in the prediction of the response of an individual patient to a defined therapeutic regimen.
  • Microarrays provide for the analysis of large amounts of genetic information, thereby providing a genetic fingerprint of an individual. There is much enthusiasm that this technology will ultimately provide the necessary tools for custom-made drug treatment regimens.
  • the invention is directed to methods of using a collection of gene product markers expressed in cancer such that when some or all of the transcripts are over or under-expressed, they identify a subtype of cancer that has a deficiency in DNA damage repair. Designation of this subtype can be considered a diagnostic test as it is not related to any specific drug but rather describes the biology of the cancer in a manner that has utility in screening and selecting appropriate cancer therapies.
  • the invention also provides methods for indicating responsiveness or resistance to DNA-damage therapeutic agents.
  • this gene or gene product list may form the basis of a single parameter or a multiparametric predictive test that could be delivered using methods known in the art such as microarray, Q-PCR, immunohistochemistry, ELISA or other technologies that can quantify mRNA or protein expression.
  • the biological pathway described herein is a feature of cancer itself, similar to grade and stage, and as such, is not limited to a single cancer disease type.
  • the collection of genes or gene products may be used to predict responsiveness of cancer therapeutics across different cancer types in different tissues. In one embodiment of the invention, these genes or gene products are useful for evaluating both breast and ovarian cancer tumors.
  • the invention described herein is not limited to any one drug; it can be used to identify responders and non responders to any of a range of drugs that directly or indirectly affect DNA damage and/or DNA damage repair e.g.
  • neoadjuvant 5-fluorouracil, anthracycline and cyclophosphamide based regimens such as FEC (5-fluorouracil/epirubicin/cyclophosphamide) and FAC (5-fluorouracil/ Adriamycin/cyclophosphamide).
  • FEC fluorouracil/epirubicin/cyclophosphamide
  • FAC fluorouracil/ Adriamycin/cyclophosphamide
  • this invention it is useful for evaluating paclitaxel, fluorouracil, doxorubicin (Adriamycin), and cyclophosphamide (T/FAC) neoadjuvant treatment in breast cancer.
  • T/FAC cyclophosphamide
  • platinum or platinum plus taxol treatment in ovarian cancer it is useful for evaluating platinum or platinum plus taxol treatment in ovarian cancer.
  • the present invention relates to prediction of response to drugs using different classifications of response, such as overall survival, progression free survival, radiological response, as defined by RECIST, complete response, partial response, stable disease and serological markers such as, but not limited to, PSA, CEA, CA125, CA15-3 and CA19-9.
  • this invention can be used to evaluate pathological complete response in breast cancer treated with FEC or FAC either alone or in the context of standard treatment, or RECIST and serum CA125 levels in ovarian cancer.
  • the present invention relates to the identification of a DNA damage response deficiency (DDRD) molecular subtype in breast and ovarian cancer.
  • DDRD DNA damage response deficiency
  • This molecular subtype can be detected by the use of two different gene classifiers - one being 40 genes in length and one being 44 genes in length.
  • the DDRD classifier was first defined by a classifier consisting of 53 probesets on the Almac Breast Disease Specific Array (DSATM). So as to validate the functional relevance of this classifier in the context of its ability to predict response to DNA-damaging containing chemotherapy regimens, the classifier needed to be re-defined at a gene level.
  • DSATM Almac Breast Disease Specific Array
  • kits for conventional diagnostic uses listed above such as qPCR, microarray, and immunoassays such as immunohistochemistry, ELISA, Western blot and the like.
  • kits include appropriate reagents and directions to assay the expression of the genes or gene products and quantify mRNA or protein expression.
  • the invention also provides methods for identifying DNA damage response-deficient
  • DDRD human tumors. It is likely that this invention can be used to identify patients that are sensitive to and respond, or are resistant to and do not respond, to drugs that damage DNA directly, damage DNA indirectly or inhibit normal DNA damage signaling and/or repair processes.
  • the invention also relates to guiding conventional treatment of patients.
  • the invention also relates to selecting patients for clinical trials where novel drugs of the classes that directly or indirectly affect DNA damage and/or DNA damage repair.
  • the present invention and methods accommodate the use of archived formalin fixed paraffin-embedded (FFPE) biopsy material, as well as fresh/frozen (FF) tissue, for assay of all transcripts in the invention, and are therefore compatible with the most widely available type of biopsy material.
  • the expression level may be determined using RNA obtained from FFPE tissue, fresh frozen tissue or fresh tissue that has been stored in solutions such as RNAlater®.
  • FIG. 1 provides a diagram representing the hierarchical analysis of ER-negative (A) and ER- positive (B) BRCAl/2 mutant and sporadic wildtype control breast samples.
  • Probeset cluster groups are annotated on the right-hand side and pathway analysis of each probeset cluster group is annotated on the left-hand side of each image.
  • the legend for each image indicates a sample's mutational status as well as the signature group each sample was assigned to for classifier generation.
  • FIG. 2 provides a diagramofbox plots comparing the AUC performance of each classification model under a 10 repeats of 5 -fold cross validation for (A) the combined sample set, (B) the ER- negative sample set and (C) the ER-positive sample set. (D) Sensitivity plus specificity plot of the cross validation predictions used to select threshold. The maximum sensitivity plus specificity is 1.682 with a corresponding signature score of -0.37.
  • FIG. 3 provides a diagram of a ROC curve of the classification performance for predicting BRCA status using the 44-gene classifier model, estimated by cross validation.
  • the AUC is -0.68 following application the classifier model.
  • the 95% confidence limits have been estimated from bootstrap with 1000 iterations.
  • FIG. 4 provides a diagram of a ROC curve of the classification performance of the 44-gene classifier model in a combined analysis of three independent datasets: FEC, FAC1 and FAC2 (Bonnefoi et al., 2007; Iwamotoet al.,] Natl Cancer Inst 103, 264-272 (2011); Lee, J. K., et al. Clin Cancer Res 16, 711-718 (2010)for predicting response to anthracyc line -based chemotherapy.
  • the AUC is -0.78 following application of the classifier model.
  • the 95% confidence limits have been estimated from bootstrap with 1000 iterations.
  • FIG. 5 provides a diagram of a ROC curve of the classification performance of the 44-gene classifier model in a combined analysis of three independent datasets in response in T/FAC treated samples(Hesset al., J Clin Oncol 24, 4236-4244 (2006); Lee et al, 2010; Tabchy, A., et a/.Clin Cancer Res 16, 5351-5361 (2010).
  • the AUC is -0.61 following application of the classifier model respectively.
  • the 95%> confidence limits were determined using 1000 bootstrap iterations.
  • FIG. 6 provides a diagram of a ROC curve of the classification performance of the 44-gene classifier model within 259 serous ovarian cancer samples in response in platinum and taxol treated samples from the in-house Almac Diagnostics ovarian dataset.
  • the AUC is -0.68 following application of the classifier model.
  • the 95% confidence limits were determined using 1000 bootstrap iterations.
  • FIG. 7 provides a histogram representation of the 44-gene DDRD classifier scores in bone marrow samples taken from healthy donors and patients with Fanconi Anaemia mutations.
  • the AUC is 0.90 following application of the classifier model.
  • the 95% confidence limits were determined using 1000 bootstrap iterations.
  • FIG. 8 provides a figure correlating the 44-gene classifier model with therapeutic response in BRCA1 mutant and wildtype cell-lines.
  • A Western blot analysis confirming increased expression of BRCA1 in the HCC1937-BR cells compared with the HCC1937-EV cells.
  • B Mean 44-gene model (DDRD) classifier score ( ⁇ SEM) within the control vector-only transfected HCC1937 (HCC1937-EV) and HCC1937 with returned exogenous expression of BRCA1 (HCC1937-BR) cell-lines. Histogram representation of cell-viability of HCC1937 parental and HCC1937-BR cells under constant exposure to a range of concentrations of PARP inhibitor KU0058948 (C) and cisplatin (D).
  • a major goal of current research efforts in cancer is to increase the efficacy of perioperative systemic therapy in patients by incorporating molecular parameters into clinical therapeutic decisions.
  • Pharmacogenetics/genomics is the study of genetic/genomic factors involved in an individual's response to a foreign compound or drug. Agents or modulators which have a stimulatory or inhibitory effect on expression of a marker of the invention can be administered to individuals to treat (prophylactically or therapeutically) cancer in a patient. It is ideal to also consider the pharmacogenomics of the individual in conjunction with such treatment. Differences in metabolism of therapeutics may possibly lead to severe toxicity or therapeutic failure by altering the relationship between dose and blood concentration of the pharmacologically active drug.
  • understanding the pharmacogenomics of an individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments.
  • Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens.
  • the level of expression of a marker of the invention in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
  • the invention is directed to a unique collection of gene or gene product markers (hereinafter referred to as "biomarkers")expressed in a cancer tissue.
  • biomarkers gene or gene product markers
  • this biomarker list may form the basis of a single parameter or multiparametric predictive test that could be delivered using methods known in the art such as microarray, Q-PCR, immunohistochemistry, ELISA or other technologies that can quantify mR A or protein expression.
  • the present invention also relates to kits and methods that are useful for prognosis following cytotoxic chemotherapy or selection of specific treatments for cancer. Methods are provided such that when some or all of the transcripts are over or under-expressed, the expression profile indicates responsiveness or resistance to DNA-damage therapeutic agents. These kits and methods employ gene or gene product markers that are differentially expressed in tumors of patients with cancer. In one embodiment of the invention, the expression profiles of these biomarkers are correlated with clinical outcome (response or survival) in archival tissue samples under a statistical method or a correlation model to create a database or model correlating expression profile with responsiveness to one or more DNA-damage therapeutic agents.
  • the predictive model may then be used to predict the responsiveness in a patient whose responsiveness to the DNA-damage therapeutic agent(s) is unknown.
  • a patient population can be divided into at least two classes based on patients' clinical outcome, prognosis, or responsiveness to DNA-damage therapeutic agents, and the biomarkers are substantially correlated with a class distinction between these classes of patients.
  • the biological pathways described herein are common to cancer as a disease, similar to grade and stage, and as such, the classifiers and methods are not limited to a single cancer disease type.
  • a unique collection of biomarkers as a genetic classifier expressed in a cancer tissue is provided that is useful in determining responsiveness or resistance to therapeutic agents, such as DNA-damage therapeutic agents, used to treat cancer.
  • Such a collection may be termed a "marker panel”, “expression classifier”, or “classifier”.
  • biomarkers useful in the present methods are identified in Table 1. These biomarkers are identified as having predictive value to determine a patient response to a therapeutic agent, or lack thereof. Their expression correlates with the response to an agent, and more specifically, a DNA-damage therapeutic agent. By examining the expression of a collection of the identified biomarkers in a tumor, it is possible to determine which therapeutic agent or combination of agents will be most likely to reduce the growth rate of a cancer, and in some embodiments, breast or ovarian cancer cells. By examining a collection of identified transcript gene or gene product markers, it is also possible to determine which therapeutic agent or combination of agents will be the least likely to reduce the growth rate of a cancer.
  • these determinations can be made on a patient-by-patient basis or on an agent-by-agent basis. Thus, one can determine whether or not a particular therapeutic regimen is likely to benefit a particular patient or type of patient, and/or whether a particular regimen should be continued.
  • biomarker panels selected from the biomarkers in Table 1 can be generated using the methods provided herein and can comprise between one, and all of the biomarkers set forth in Table 1 and each and every combination in between (e.g., four selected biomarkers, 16 selected biomarkers, 74 selected biomarkers, etc.).
  • the predictive biomarker set comprises at least 5, 10, 20, 40, 60, 100, 150, 200, or 300 or more biomarkers. In other embodiments, the predictive biomarker set comprises no more than 5, 10, 20, 40, 60, 100, 150, 200, 300, 400, 500, 600 or 700 biomarkers.
  • the predictive biomarker set includes a plurality of biomarkers listed in Table 1. In some embodiments the predictive biomarker set includes at least about 1%, about 5%, about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%), about 96%>, about 97%, about 98%>, or about 99% of the biomarkers listed in Table 1. Selected predictive biomarker sets can be assembled from the predictive biomarkers provided using methods described herein and analogous methods known in the art. In one embodiment, the biomarker panel contains all 203 biomarkers in Table 1. In another embodiment, the biomarker panel contains 40 or 44 biomarkers in Table 1 or 2.
  • Predictive biomarker sets may be defined in combination with corresponding scalar weights on the real scale with varying magnitude, which are further combined through linear or non-linear, algebraic, trigonometric or correlative means into a single scalar value via an algebraic, statistical learning, Bayesian, regression, or similar algorithms which together with a mathematically derived decision function on the scalar value provide a predictive model by which expression profiles from samples may be resolved into discrete classes of responder or non-responder, resistant or non-resistant, to a specified drug or drug class.
  • Such predictive models are developed by learning weights and the decision threshold, optimized for sensitivity, specificity, negative and positive predictive values, hazard ratio or any combination thereof, under cross-validation, bootstrapping or similar sampling techniques, from a set of representative expression profiles from historical patient samples with known drug response and/or resistance.
  • the biomarkers are used to form a weighted sum of their signals, where individual weights can be positive or negative.
  • the resulting sum (“decisive function") is compared with a pre-determined reference point or value. The comparison with the reference point or value may be used to diagnose, or predict a clinical condition or outcome.
  • biomarkers included in the classifier provided in Table 1 will carry unequal weights in a classifier for responsiveness or resistance to a therapeutic agent. Therefore, while as few as one sequence may be used to diagnose or predict an outcome such as responsiveness to therapeutic agent, the specificity and sensitivity or diagnosis or prediction accuracy may increase using more sequences.
  • weight refers to the relative importance of an item in a statistical calculation.
  • the weight of each biomarker in a gene expression classifier may be determined on a data set of patient samples using analytical methods known in the art.
  • the biomarker panel is directed to the 40 biomarkers detailed in Table 2A with corresponding ranks and weights detailed in the table or alternative rankings and weightings, depending, for example, on the disease setting.
  • the biomarker panel is directed to the 44 biomarkers detailed in Table 2B with corresponding ranks and weights detailed in the table or alternative rankings and weightings, depending, for example, on the disease setting.
  • Tables 2A and 2B rank the biomarkers in order of decreasing weight in the classifier, defined as the rank of the average weight in the compound decision score function measured under cross-validation.
  • Table 2C present the probe sets that represent the genes in Table 2 A and 2B with reference to their sequence ID numbers.
  • Table 2D presents the antisense probe sequences that were present on the array for the genes in the signatures.
  • subsets of the biomarkers listed in Table 2A and Table 2B may be used in the methods described herein. These subsets include but are not limited to biomarkers ranked 1-2, 1-3, 1-4, 1-5, 1-10, 1-20, 1-30, 1-40, 1-44, 6-10, 11-15, 16-20, 21-25, 26-30, 31-35, 36-40, 36-44, 11-20, 21-30, 31-40, and 31-44 in Table 2A or Table 2B.
  • therapeutic responsiveness is predicted in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to at least one of the biomarkers GBP5, CXCL10, IDOl and MX1 and at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, or 36.
  • biomarker can refer to a gene, an mRNA, cDNA, an antisense transcript, a miRNA, a polypeptide, a protein, a protein fragment, or any other nucleic acid sequence or polypeptide sequence that indicates either gene expression levels or protein production levels.
  • the biomarker comprises an mRNA of CXCL10, IDOl, CD2, GBP5, PRAME, ITGAL, LRP4, APOL3, CDR1, FYB, TSPAN7, RAC2, KLHDC7B, GRB14, AC138128.1, KIF26A, CD274, ETV7, MFAP5, OLFM4, PI15, FOSB, FAM19A5, NLRC5, PRICKLEl, EGRl, CLDNIO, ADAMTS4, SP140L, ANXAl, RSAD2, ESRl, IKZF3, OR2I1P, EGFR, NAT1, LATS2, CYP2B6, PTPRC, PPP1R1A, or AL137218.1
  • the biomarker comprises an mRNA of CXCL10, IDOl, CD2, GBP5, PRAME, ITGAL, LRP4, APOL3, CDR1, FYB, TSPAN7, RAC2, KLHDC7B, GRB14, AC138128.1,
  • the biomarker comprises an antisense transcript of MXl, IFI44L, GBP5, BIRC3, IGJ, IQGAP3, LOC100294459, SIXl, SLC9A3R1, STAT1, TOB1, UBD, C1QC, C2orfl4, EPSTI, GALNT6, HIST1H4H, HIST2H4B, KIAA1244, LOCI 00287927, LOCI 00291682, or LOCI 00293679
  • the biomarker comprises an antisense transcript of MXl, IFI44L, GBP5, BIRC3, IGJ, IQGAP3, LOC100294459, SIXl, SLC9A3R1, STAT1, TOB1, UBD, C1QC, C2orfl4, EPSTI, GALNT6, HIST1H4H, HIST2H4B, KIAA1244, LOCI 00287927, LOCI 00291682, or LOCI 0029
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarkers GBP5, CXCLIO, IDOl and MXl and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, or 36.
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker GBP5 and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 29, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38 or 39.
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker CXCLIO and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 29, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38 or 39.
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker IDOl and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 29, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38 or 39.
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker MX-1 and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 29, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38 or 39.
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to at least two of the biomarkers CXCLIO, MXl, IDOl and IFI44L and at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40.
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarkers CXCLIO, MXl, IDOl and IFI44L and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27,
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker CXCLIO and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 29, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42 or 43.
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker MXl and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 29, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42 or 43.
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker IDOl and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18,
  • therapeutic responsiveness is predicted, or a cancer diagnosis is indicated, in an individual by conducting an assay on a biological sample from the individual and detecting biomarker values that each correspond to the biomarker IFI44L and one of at least N additional biomarkers selected from the list of biomarkers in Table 2B, wherein N equals 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 29, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42 or 43.
  • the probes listed in Table 2C may be used in the methods described herein. These subsets include but are not limited to a subset of SEQ ID NOs corresponding to one or more of GBP5, CXCL10, IDOl, MX1, IF 1441, CD2, PRAME, ITGAL, LRP4, and APOL3.
  • the probes correspond to all of the biomarkers CXCL10, MX1, IDOl, IF144L, CD2, GBP5, PRAME, ITGAL, LRP4, APOL3, CDR1, FYB, TSPAN7, RAC2, KLHDC7B, GRB14, AC 138128.1, KIF26A, CD274, CD 109, ETV7, MFAP5, OLFM4, PI 15, FOSB, FAM19A5, NLRC5, PRICKLEl, EGR1, CLDN10, ADAMTS4, SP140L, ANXAl, RSAD2, ESR1, IKZF3, OR211P, EGFR, NAT1, LATS2, CYP2B6, PTPRC, PPP1R1A, and AL137218.1.
  • each subset can include multiple probes directed to the same biomarker.
  • the probes represented by SEQ ID NOs: 135, 140, 142 and 195 are all directed to GBP5.
  • a subset containing probes directed or corresponding to GBP5 includes one or more of SEQ ID NOs: 135, 140, 142 and 195.
  • a subset containing probes directed to or corresponding to CXCL10 includes one or more of SEQ ID NOs: 131 and 160.
  • biomarkers and diagnose disease A variety of methods have been utilized in an attempt to identify biomarkers and diagnose disease.
  • protein-based markers these include two-dimensional electrophoresis, mass spectrometry, and immunoassay methods.
  • nucleic acid markers these include mRNA expression profiles, microRNA profiles, FISH, serial analysis of gene expression (SAGE), methylation profiles, and large-scale gene expression arrays.
  • biomarker When a biomarker indicates or is a sign of an abnormal process, disease or other condition in an individual, that biomarker is generally described as being either over-expressed or under-expressed as compared to an expression level or value of the biomarker that indicates or is a sign of a normal process, an absence of a disease or other condition in an individual.
  • Up- regulation “up-regulated”, “over-expression”, “over-expressed”, and any variations thereof are used interchangeably to refer to a value or level of a biomarker in a biological sample that is greater than a value or level (or range of values or levels) of the biomarker that is typically detected in similar biological samples from healthy or normal individuals.
  • the terms may also refer to a value or level of a biomarker in a biological sample that is greater than a value or level (or range of values or levels) of the biomarker that may be detected at a different stage of a particular disease.
  • Down-regulation Down-regulated
  • under-expression under-expressed
  • any variations thereof are used interchangeably to refer to a value or level of a biomarker in a biological sample that is less than a value or level (or range of values or levels) of the biomarker that is typically detected in similar biological samples from healthy or normal individuals.
  • the terms may also refer to a value or level of a biomarker in a biological sample that is less than a value or level (or range of values or levels) of the biomarker that may be detected at a different stage of a particular disease.
  • a biomarker that is either over-expressed or under-expressed can also be referred to as being “differentially expressed” or as having a “differential level” or “differential value” as compared to a "normal” expression level or value of the biomarker that indicates or is a sign of a normal process or an absence of a disease or other condition in an individual.
  • "differential expression” of a biomarker can also be referred to as a variation from a "normal” expression level of the biomarker.
  • differential biomarker expression and “differential expression” are used interchangeably to refer to a biomarker whose expression is activated to a higher or lower level in a subject suffering from a specific disease, relative to its expression in a normal subject, or relative to its expression in a patient that responds differently to a particular therapy or has a different prognosis.
  • the terms also include biomarkers whose expression is activated to a higher or lower level at different stages of the same disease. It is also understood that a differentially expressed biomarker may be either activated or inhibited at the nucleic acid level or protein level, or may be subject to alternative splicing to result in a different polypeptide product.
  • Differential biomarker expression may include a comparison of expression between two or more genes or their gene products; or a comparison of the ratios of the expression between two or more genes or their gene products; or even a comparison of two differently processed products of the same gene, which differ between normal subjects and subjects suffering from a disease; or between various stages of the same disease.
  • Differential expression includes both quantitative, as well as qualitative, differences in the temporal or cellular expression pattern in a biomarker among, for example, normal and diseased cells, or among cells which have undergone different disease events or disease stages.
  • the expression profile obtained is a genomic or nucleic acid expression profile, where the amount or level of one or more nucleic acids in the sample is determined.
  • the sample that is assayed to generate the expression profile employed in the diagnostic or prognostic methods is one that is a nucleic acid sample.
  • the nucleic acid sample includes a population of nucleic acids that includes the expression information of the phenotype determinative biomarkers of the cell or tissue being analyzed.
  • the nucleic acid may include RNA or DNA nucleic acids, e.g., mR A, cRNA, cDNA etc., so long as the sample retains the expression information of the host cell or tissue from which it is obtained.
  • the sample may be prepared in a number of different ways, as is known in the art, e.g., by mRNA isolation from a cell, where the isolated mRNA is used as isolated, amplified, or employed to prepare cDNA, cRNA, etc., as is known in the field of differential gene expression. Accordingly, determining the level of mRNA in a sample includes preparing cDNA or cRNA from the mRNA and subsequently measuring the cDNA or cRNA.
  • the sample is typically prepared from a cell or tissue harvested from a subject in need of treatment, e.g., via biopsy of tissue, using standard protocols, where cell types or tissues from which such nucleic acids may be generated include any tissue in which the expression pattern of the to be determined phenotype exists, including, but not limited to, disease cells or tissue, body fluids, etc.
  • the expression profile may be generated from the initial nucleic acid sample using any convenient protocol. While a variety of different manners of generating expression profiles are known, such as those employed in the field of differential gene expression/biomarker analysis, one representative and convenient type of protocol for generating expression profiles is array- based gene expression profile generation protocols. Such applications are hybridization assays in which a nucleic acid that displays "probe" nucleic acids for each of the genes to be assayed/profiled in the profile to be generated is employed. In these assays, a sample of target nucleic acids is first prepared from the initial nucleic acid sample being assayed, where preparation may include labeling of the target nucleic acids with a label, e.g., a member of a signal producing system.
  • a label e.g., a member of a signal producing system.
  • target nucleic acid sample preparation Following target nucleic acid sample preparation, the sample is contacted with the array under hybridization conditions, whereby complexes are formed between target nucleic acids that are complementary to probe sequences attached to the array surface. The presence of hybridized complexes is then detected, either qualitatively or quantitatively.
  • Specific hybridization technology which may be practiced to generate the expression profiles employed in the subject methods includes the technology described in U.S. Pat. Nos.
  • the relative expression levels of biomarkers in a cancer tissue are measured to form a gene expression profile.
  • the gene expression profile of a set of biomarkers from a patient tissue sample is summarized in the form of a compound decision score and compared to a score threshold that is mathematically derived from a training set of patient data.
  • the score threshold separates a patient group based on different characteristics such as, but not limited to, responsiveness/non-responsiveness to treatment.
  • the patient training set data is preferably derived from cancer tissue samples having been characterized by prognosis, likelihood of recurrence, long term survival, clinical outcome, treatment response, diagnosis, cancer classification, or personalized genomics profile.
  • Expression profiles, and corresponding decision scores from patient samples may be correlated with the characteristics of patient samples in the training set that are on the same side of the mathematically derived score decision threshold.
  • the threshold of the linear classifier scalar output is optimized to maximize the sum of sensitivity and specificity under cross-validation as observed within the training dataset.
  • the overall expression data for a given sample is normalized using methods known to those skilled in the art in order to correct for differing amounts of starting material, varying efficiencies of the extraction and amplification reactions, etc.
  • Using a linear classifier on the normalized data to make a diagnostic or prognostic call effectively means to split the data space, i.e. all possible combinations of expression values for all genes in the classifier, into two disjoint halves by means of a separating hyperplane. This split is empirically derived on a large set of training examples, for example from patients showing responsiveness or resistance to a therapeutic agent.
  • the biomarker expression profile of a patient tissue sample is evaluated by a linear classifier.
  • a linear classifier refers to a weighted sum of the individual biomarker intensities into a compound decision score ("decision function"). The decision score is then compared to a pre-defined cut-off score threshold, corresponding to a certain set-point in terms of sensitivity and specificity which indicates if a sample is above the score threshold (decision function positive) or below (decision function negative).
  • the data space i.e. the set of all possible combinations of biomarker expression values
  • the data space is split into two mutually exclusive halves corresponding to different clinical classifications or predictions, e.g. one corresponding to responsiveness to a therapeutic agent and the other to resistance.
  • relative over-expression of a certain biomarker can either increase the decision score (positive weight) or reduce it (negative weight) and thus contribute to an overall decision of, for example, responsiveness or resistance to a therapeutic agent.
  • AUC area under the curve
  • ROC receiver operating characteristic
  • the feature data across the entire population e.g., the cases and controls
  • the true positive and false positive rates for the data are calculated.
  • the true positive rate is determined by counting the number of cases above the value for that feature and then dividing by the total number of cases.
  • the false positive rate is determined by counting the number of controls above the value for that feature and then dividing by the total number of controls.
  • ROC curves can be generated for a single feature as well as for other single outputs, for example, a combination of two or more features can be mathematically combined (e.g., added, subtracted, multiplied, etc.) to provide a single sum value, and this single sum value can be plotted in a ROC curve. Additionally, any combination of multiple features, in which the combination derives a single output value, can be plotted in a ROC curve. These combinations of features may comprise a test.
  • the ROC curve is the plot of the true positive rate (sensitivity) of a test against the false positive rate (1 -specificity) of the test.
  • this quantity i.e. the cut-off threshold responsiveness or resistance to a therapeutic agent
  • the interpretation of this quantity is derived in the development phase ("training") from a set of patients with known outcome.
  • the corresponding weights and the responsiveness/resistance cut-off threshold for the decision score are fixed a priori from training data by methods known to those skilled in the art.
  • Partial Least Squares Discriminant Analysis (PLS-DA) is used for determining the weights.
  • PLS-DA Partial Least Squares Discriminant Analysis
  • Other methods for performing the classification known to those skilled in the art, may also be with the methods described herein when applied to the transcripts of a cancer classifier.
  • a training step a set of patient samples for both responsiveness/resistance cases are measured and the prediction method is optimised using the inherent information from this training data to optimally predict the training set or a future sample set.
  • the used method is trained or parameterised to predict from a specific intensity pattern to a specific predictive call. Suitable transformation or pre-processing steps might be performed with the measured data before it is subjected to the prognostic method or algorithm.
  • a weighted sum of the pre-processed intensity values for each transcript is formed and compared with a threshold value optimised on the training set (Duda et al. Pattern Classification, 2 nd ed., John Wiley, New York 2001).
  • the weights can be derived by a multitude of linear classification methods, including but not limited to Partial Least Squares (PLS, (Nguyen et al, 2002, Bioinformatics 18 (2002) 39-50)) or Support Vector Machines (SVM, (Scholkopf et al. Learning with Kernels, MIT Press, Cambridge 2002)).
  • the data is transformed non-linearly before applying a weighted sum as described above.
  • This non-linear transformation might include increasing the dimensionality of the data.
  • the non-linear transformation and weighted summation might also be performed implicitly, e.g. through the use of a kernel function. (Scholkopf et al. Learning with Kernels, MIT Press, Cambridge 2002).
  • a new data sample is compared with two or more class prototypes, being either real measured training samples or artificially created prototypes.
  • This comparison is performed using suitable similarity measures, for example, but not limited to Euclidean distance (Duda et al. Pattern Classification, 2 nd ed., John Wiley, New York 2001), correlation coefficient (Van't Veer, et al. 2002, Nature 415:530) etc.
  • a new sample is then assigned to the prognostic group with the closest prototype or the highest number of prototypes in the vicinity.
  • decision trees (Hastie et al., The Elements of
  • neural networks (Bishop, Neural Networks for Pattern Recognition, Clarendon Press, Oxford 1995) are used to make a prognostic call from the measured intensity data for the transcript set or their products.
  • discriminant analysis (Duda et al., Pattern Classification, 2 nd ed., John Wiley, New York 2001), comprising but not limited to linear, diagonal linear, quadratic and logistic discriminant analysis, is used to make a prognostic call from the measured intensity data for the transcript set or their products.
  • PAM Prediction Analysis for Microarrays
  • Soft Independent Modelling of Class Analogy (SIMCA, (Wold, 1976, Pattern Recogn. 8:127-139)) is used to make a predictive call from the measured intensity data for the transcript set or their products.
  • DNA-damage therapeuticagent includes agents known to damage DNA directly, agents that prevent DNA damage repair, agents that inhibit DNA damage signaling, agents that inhibit DNA damage induced cell cycle arrest, and agents that inhibit processes indirectly leading to DNA damage.
  • DNA-damage therapeuticagent includes agents known to damage DNA directly, agents that prevent DNA damage repair, agents that inhibit DNA damage signaling, agents that inhibit DNA damage induced cell cycle arrest, and agents that inhibit processes indirectly leading to DNA damage.
  • Alkylating agents platinum containing agents such as cisplatin, carboplatin, and oxaliplatin; cyclophosphamide; busulphan).
  • Topisomerase II inhibitors etoposide;anthracylcines such as doxorubicin and epirubicin
  • Inhibitors of base excision repair (PARP inhibitors, AGO 14699, AZD2281 , ABT- 888, MK4827, BSI-201, INO-1001, TRC-102, APEX 1 inhibitors, APEX 2 inhibitors, Ligase III inhibitors
  • CHK 1 inhibitors (XL-844,UCN-01, AZD7762, PF00477736)
  • CHK 2 inhibitors (XL-844, AZD7762, PF00477736)
  • the predictive classifiers described herein are useful for determining responsiveness or resistance to a therapeutic agentfor treating cancer.
  • the biological pathway described herein is a feature of cancer itself, similar to grade and stage, and as such, is not limited to a single cancer disease type. Therefore, the collection of genes or gene products may be used to predict responsiveness of cancer therapeutics across different cancer types in different tissues. In one embodiment, this collection of genes or gene products is useful for evaluating both breast and ovarian cancer tumors.
  • cancer includes, but is not limited to, leukemia, brain cancer, prostate cancer, liver cancer, ovarian cancer, stomach cancer, colorectal cancer, throat cancer, breast cancer, skin cancer, melanoma, lung cancer, sarcoma, cervical cancer, testicular cancer, bladder cancer, endocrine cancer, endometrial cancer, esophageal cancer, glioma, lymphoma, neuroblastoma, osteosarcoma, pancreatic cancer, pituitary cancer, renal cancer, and the like.
  • the methods described herein refer to cancers that are treated with chemotherapeutic agents of the classes DNA damaging agents, DNA repair target therapies, inhibitors of DNA damage signalling, inhibitors of DNA damage induced cell cycle arrest and inhibition of processes indirectly leading to DNA damage, but not limited to these classes.
  • chemotherapeutic agents of the classes DNA damaging agents, DNA repair target therapies, inhibitors of DNA damage signalling, inhibitors of DNA damage induced cell cycle arrest and inhibition of processes indirectly leading to DNA damage, but not limited to these classes.
  • Each of these chemotherapeutic agents is considered a "DNA-damage therapeutic agent" as the term is used herein.
  • Bio sample “sample”, and “test sample” are used interchangeably herein to refer to any material, biological fluid, tissue, or cell obtained or otherwise derived from an individual.
  • a blood sample can be fractionated into serum or into fractions containing particular types of blood cells, such as red blood cells or white blood cells (leukocytes).
  • a sample can be a combination of samples from an individual, such as a combination of a tissue and fluid sample.
  • biological sample also includes materials containing homogenized solid material, such as from a stool sample, a tissue sample, or a tissue biopsy, for example.
  • biological sample also includes materials derived from a tissue culture or a cell culture.
  • any suitable methods for obtaining a biological sample can be employed; exemplary methods include, e.g., phlebotomy, swab (e.g., buccal swab), and a fine needle aspirate biopsy procedure. Samples can also be collected, e.g., by micro dissection (e.g., laser capture micro dissection (LCM) or laser micro dissection (LMD)), bladder wash, smear (e.g., a PAP smear), or ductal lavage.
  • a "biological sample” obtained or derived from an individual includes any such sample that has been processed in any suitable manner after being obtained from the individual.
  • the target cells may be tumor cells, for example colon cancer cells or stomach cancer cells.
  • the target cells are derived from any tissue source, including human and animal tissue, such as, but not limited to, a newly obtained sample, a frozen sample, a biopsy sample, a sample of bodily fluid, a blood sample, preserved tissue such as a paraffin-embedded fixed tissue sample (i.e., a tissue block), or cell culture.
  • Kits for Gene Expression Analysis can be provided in a kit.
  • the kit can contain reagents, tools, and instructions for determining an appropriate therapy for a cancer patient.
  • a kit can include reagents for collecting a tissue sample from a patient, such as by biopsy, and reagents for processing the tissue.
  • the kit can also include one or more reagents for performing a biomarker expression analysis, such as reagents for performing RT-PCR, qPCR, northern blot, proteomic analysis, or immunohistochemistry to determine expression levels of biomarkers in a sample of a patient.
  • primers for performing RT-PCR can be included in such kits.
  • Appropriate buffers for the assays can also be included.
  • Detection reagents required for any of these assays can also be included. The appropriate reagents and methods are described in further detail below.
  • kits featured herein can also include an instruction sheet describing how to perform the assays for measuring biomarker expression.
  • the instruction sheet can also include instructions for how to determine a reference cohort, including how to determine expression levels of biomarkers in the reference cohort and how to assemble the expression data to establish a reference for comparison to a test patient.
  • the instruction sheet can also include instructions for assaying biomarker expression in a test patient and for comparing the expression level with the expression in the reference cohort to subsequently determine the appropriate chemotherapy for the test patient. Methods for determining the appropriate chemotherapy are described above and can be described in detail in the instruction sheet.
  • kits can be descriptive, instructional, marketing or other material that relates to the methods described herein and/or the use of the reagents for the methods described herein.
  • the informational material of the kit can contain contact information, e.g., a physical address, email address, website, or telephone number, where a user of the kit can obtain substantive information about performing a gene expression analysis and interpreting the results, particularly as they apply to a human's likelihood of having a positive response to a specific therapeutic agent.
  • kits featured herein can also contain software necessary to infer a patient's likelihood of having a positive response to a specific therapeutic agent from the biomarker expression.
  • Measuring mRNA in a biological sample may be used as a surrogate for detection of the level of the corresponding protein in the biological sample.
  • any of the biomarkers or biomarker panels described herein can also be detected by detecting the appropriate RNA.
  • Methods of gene expression profiling include, but are not limited to, microarray, RT-PCT, qPCR, northern blots, SAGE, mass spectrometry.
  • mRNA expression levels are measured by reverse transcription quantitative polymerase chain reaction (RT-PCR followed with qPCR).
  • RT-PCR is used to create a cDNA from the mRNA.
  • the cDNA may be used in a qPCR assay to produce fluorescence as the DNA amplification process progresses. By comparison to a standard curve, qPCR can produce an absolute measurement such as number of copies of mRNA per cell.
  • Northern blots, microarrays, Invader assays, and RT-PCR combined with capillary electrophoresis have all been used to measure expression levels of mRNA in a sample. See Gene Expression Profiling: Methods and Protocols, Richard A. Shimkets, editor, Humana Press, 2004.
  • miRNA molecules are small RNAs that are non-coding but may regulate gene expression. Any of the methods suited to the measurement of mRNA expression levels can also be used for the corresponding miRNA. Recently many laboratories have investigated the use of miRNAs as biomarkers for disease. Many diseases involve widespread transcriptional regulation, and it is not surprising that miR As might find a role as biomarkers. The connection between miR A concentrations and disease is often even less clear than the connections between protein levels and disease, yet the value of miRNA biomarkers might be substantial.
  • RNA biomarkers have similar requirements, although many potential protein biomarkers are secreted intentionally at the site of pathology and function, during disease, in a paracrine fashion. Many potential protein biomarkers are designed to function outside the cells within which those proteins are synthesized.
  • Gene expression may also be evaluated using mass spectrometry methods.
  • a variety of configurations of mass spectrometers can be used to detect biomarker values.
  • Several types of mass spectrometers are available or can be produced with various configurations.
  • a mass spectrometer has the following major components: a sample inlet, an ion source, a mass analyzer, a detector, a vacuum system, and instrument-control system, and a data system. Difference in the sample inlet, ion source, and mass analyzer generally define the type of instrument and its capabilities.
  • an inlet can be a capillary-column liquid chromatography source or can be a direct probe or stage such as used in matrix-assisted laser desorption.
  • Common ion sources are, for example, electrospray, including nanospray and microspray or matrix-assisted laser desorption.
  • Common mass analyzers include a quadrupole mass filter, ion trap mass analyzer and time-of-flight mass analyzer. Additional mass spectrometry methods are well known in the art (see Burlingame et al, Anal. Chem. 70:647 R- 716R (1998); Kinter and Sherman, New York (2000)).
  • Protein biomarkers and biomarker values can be detected and measured by any of the following: electrospray ionization mass spectrometry (ESI-MS), ESI-MS/MS, ESI-MS/(MS)n, matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS), surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF- MS), desorption/ionization on silicon (DIOS), secondary ion mass spectrometry (SIMS), quadrupole time-of-flight (Q-TOF), tandem time-of-flight (TOF/TOF) technology, called ultraflex III TOF/TOF, atmospheric pressure chemical ionization mass spectrometry (APCI- MS), APCI-MS/MS, APCI-(MS).sup.N, atmospheric pressure photoionization mass spectrometry (APPI-MS), APPI-MS
  • Labeling methods include but are not limited to isobaric tag for relative and absolute quantitation (iTRAQ) and stable isotope labeling with amino acids in cell culture (SILAC).
  • Capture reagents used to selectively enrich samples for candidate biomarker proteins prior to mass spectroscopic analysis include but are not limited to aptamers, antibodies, nucleic acid probes, chimeras, small molecules, an F(ab') 2 fragment, a single chain antibody fragment, an Fv fragment, a single chain Fv fragment, a nucleic acid, a lectin, a ligand-binding receptor, affybodies, nanobodies, ankyrins, domain antibodies, alternative antibody scaffolds (e.g.
  • the foregoing assays enable the detection of biomarker values that are useful in methods for predicting responsiveness of a cancer therapeutic agent, where the methods comprise detecting, in a biological sample from an individual, at least N biomarker values that each correspond to a biomarker selected from the group consisting of the biomarkers provided in Tables 1 or 2, wherein a classification, as described in detail below, using the biomarker values indicates whether the individual will be responsive to a therapeutic agent. While certain of the described predictive biomarkers are useful alone for predicting responsiveness to a therapeutic agent, methods are also described herein for the grouping of multiple subsets of the biomarkers that are each useful as a panel of two or more biomarkers.
  • N can be selected to be any number from any of the above-described ranges, as well as similar, but higher order, ranges.
  • biomarker values can be detected and classified individually or they can be detected and classified collectively, as for example in a multiplex assay format.
  • the present invention makes use of "oligonucleotide arrays" (also called herein "microarrays"). Microarrays can be employed for analyzing the expression of biomarkers in a cell, and especially for measuring the expression of biomarkers of cancer tissues.
  • biomarker arrays are produced by hybridizing detectably labeled polynucleotides representing the mR A transcripts present in a cell (e.g., fluorescently-labeled cDNA synthesized from total cell mRNA or labeled cR A) to a microarray.
  • a microarray is a surface with an ordered array of binding (e.g., hybridization) sites for products of many of the genes in the genome of a cell or organism, preferably most or almost all of the genes.
  • Microarrays can be made in a number of ways known in the art. However produced, microarrays share certain characteristics. The arrays are reproducible, allowing multiple copies of a given array to be produced and easily compared with each other.
  • the microarrays are small, usually smaller than 5 cm 2 , and they are made from materials that are stable under binding (e.g., nucleic acid hybridization) conditions.
  • a given binding site or unique set of binding sites in the microarray will specifically bind the product of a single gene in the cell.
  • positionally addressable arrays containing affixed nucleic acids of known sequence at each location are used.
  • cDNA or cRNA complementary to the total cellular mRNA when detectably labeled (e.g., with a fiuorophore) cDNA or cRNA complementary to the total cellular mRNA is hybridized to a microarray, the site on the array corresponding to a gene (i.e., capable of specifically binding the product of the gene) that is not transcribed in the cell will have little or no signal (e.g., fluorescent signal), and a gene for which the encoded mRNA is prevalent will have a relatively strong signal.
  • a gene i.e., capable of specifically binding the product of the gene
  • Nucleic acid hybridization and wash conditions are chosen so that the probe "specifically binds" or “specifically hybridizes' to a specific array site, i.e., the probe hybridizes, duplexes or binds to a sequence array site with a complementary nucleic acid sequence but does not hybridize to a site with a non-complementary nucleic acid sequence.
  • one polynucleotide sequence is considered complementary to another when, if the shorter of the polynucleotides is less than or equal to 25 bases, there are no mismatches using standard base- pairing rules or, if the shorter of the polynucleotides is longer than 25 bases, there is no more than a 5% mismatch.
  • the polynucleotides are perfectly complementary (no mismatches). It can be demonstrated that specific hybridization conditions result in specific hybridization by carrying out a hybridization assay including negative controls using routine experimentation.
  • Optimal hybridization conditions will depend on the length (e.g., oligomer vs. polynucleotide greater than 200 bases) and type (e.g., RNA, DNA, PNA) of labeled probe and immobilized polynucleotide or oligonucleotide.
  • length e.g., oligomer vs. polynucleotide greater than 200 bases
  • type e.g., RNA, DNA, PNA
  • cDNA microarrays When the cDNA microarrays are used, typical hybridization conditions are hybridization in 5xSSC plus 0.2% SDS at 65C for 4 hours followed by washes at 25°C in low stringency wash buffer (lxSSC plus 0.2% SDS) followed by 10 minutes at 25°C in high stringency wash buffer (0.1SSC plus 0.2% SDS) (see Shena et al, Proc. Natl. Acad. Sci. USA, Vol. 93, p. 10614 (1996)).
  • Useful hybridization conditions are also provided in, e.g., Tijessen, Hybridization With Nucleic Acid Probes", Elsevier Science Publishers B.V. (1993) and Kricka, "Nonisotopic DNA Probe Techniques", Academic Press, San Diego, Calif. (1992).
  • Immunoassay methods are based on the reaction of an antibody to its corresponding target or analyte and can detect the analyte in a sample depending on the specific assay format.
  • monoclonal antibodies are often used because of their specific epitope recognition.
  • Polyclonal antibodies have also been successfully used in various immunoassays because of their increased affinity for the target as compared to monoclonal antibodies
  • Immunoassays have been designed for use with a wide range of biological sample matrices
  • Immunoassay formats have been designed to provide qualitative, semi-quantitative, and quantitative results.
  • Quantitative results may be generated through the use of a standard curve created with known concentrations of the specific analyte to be detected.
  • the response or signal from an unknown sample is plotted onto the standard curve, and a quantity or value corresponding to the target in the unknown sample is established.
  • ELISA or EIA can be quantitative for the detection of an analyte/biomarker. This method relies on attachment of a label to either the analyte or the antibody and the label component includes, either directly or indirectly, an enzyme. ELISA tests may be formatted for direct, indirect, competitive, or sandwich detection of the analyte. Other methods rely on labels such as, for example, radioisotopes (I 125 ) or fluorescence.
  • Additional techniques include, for example, agglutination, nephelometry, turbidimetry, Western blot, immunoprecipitation, immunocytochemistry, immunohistochemistry, flow cytometry, Luminex assay, and others (see ImmunoAssay: A Practical Guide, edited by Brian Law, published by Taylor & Francis, Ltd., 2005 edition).
  • Exemplary assay formats include enzyme-linked immunosorbent assay (ELISA), radioimmunoassay, fluorescent, chemiluminescence, and fluorescence resonance energy transfer (FRET) or time resolved-FRET (TR-FRET) immunoassays.
  • ELISA enzyme-linked immunosorbent assay
  • FRET fluorescence resonance energy transfer
  • TR-FRET time resolved-FRET
  • biomarkers include biomarker immunoprecipitation followed by quantitative methods that allow size and peptide level discrimination, such as gel electrophoresis, capillary electrophoresis, planar electrochromatography, and the like.
  • Methods of detecting and/or quantifying a detectable label or signal generating material depend on the nature of the label.
  • the products of reactions catalyzed by appropriate enzymes can be, without limitation, fluorescent, luminescent, or radioactive or they may absorb visible or ultraviolet light.
  • detectors suitable for detecting such detectable labels include, without limitation, x-ray film, radioactivity counters, scintillation counters, spectrophotometers, colorimeters, fluorometers, luminometers, and densitometers.
  • Any of the methods for detection can be performed in any format that allows for any suitable preparation, processing, and analysis of the reactions. This can be, for example, in multi-well assay plates (e.g., 96 wells or 384 wells) or using any suitable array or microarray. Stock solutions for various agents can be made manually or robotically, and all subsequent pipetting, diluting, mixing, distribution, washing, incubating, sample readout, data collection and analysis can be done robotically using commercially available analysis software, robotics, and detection instrumentation capable of detecting a detectable label.
  • methods are provided for identifying and/or selecting a cancer patient who is responsive to a therapeutic regimen.
  • the methods are directed to identifying or selecting a cancer patient who is responsive to a therapeutic regimen that includes administering an agent that directly or indirectly damages DNA.
  • Methods are also provided for identifying a patient who is non-responsive to a therapeutic regimen.
  • These methods typically include determining the level of expression of a collection of predictive markers in a patient's tumor (primary, metastatic or other derivatives from the tumor such as, but not limited to, blood, or components in blood, urine, saliva and other bodily fluids)(e.g., a patient's cancer cells), comparing the level of expression to a reference expression level, and identifying whether expression in the sample includes a pattern or profile of expression of a selected predictive biomarker or biomarker set which corresponds to response or non-response to therapeutic agent.
  • a patient's tumor primary, metastatic or other derivatives from the tumor such as, but not limited to, blood, or components in blood, urine, saliva and other bodily fluids
  • a patient's cancer cells e.g., a patient's cancer cells
  • a method of predicting responsiveness of an individual to a DNA- damage therapeutic agent comprises the following steps: obtaining a test sample from the individual; measuring expression levels of one or more biomarkers in the test sample, wherein the one or more biomarkers are selected from the group consisting of CXCL10, MX1, IDOl, IF144L, CD2, GBP5, PRAME, ITGAL, LRP4, and APOL3; deriving a test score that captures the expression levels; providing a threshold score comprising information correlating the test score and responsiveness; and comparing the test score to the threshold score; wherein responsiveness is predicted when the test score exceeds the threshold score.
  • One of ordinary skill in the art can determine an appropriate threshold score, and appropriate biomarker weightings, using the teachings provided herein including the teachings of Example 1.
  • the method of predicting responsiveness of an individual to a DNA-damage therapeutic agent comprises measuring the expression levels of one or more biomarkers in the test sample, wherein the one or more biomarkers are selected from the group consisting of CXCL10, MX1, IDOl, IF144L, CD2, GBP5, PRAME, ITGAL, LRP4, APOL3, CDR1, FYB, TSPAN7, RAC2, KLHDC7B, GRB14, AC138128.1, KIF26A, CD274, CD109, ETV7, MFAP5, OLFM4, PI15, FOSB, FAM19A5, NLRC5, PRICKLEl, EGR1, CLDN10, ADAMTS4, SP140L, ANXA1, RSAD2, ESR1, IKZF3, OR211P, EGFR, NAT1, LATS2, CYP2B6, PTPRC, PPP1R1A, and AL137218.1.
  • the one or more biomarkers are selected from the group consisting of CXCL
  • Tables 2A and 2B provide exemplary gene signatures (or gene classifiers) wherein the biomarkers consist of 40 or 44 of the gene products listed therein, respectively, and wherein a threshold score is derived from the individual gene product weightings listed therein.
  • a test score that exceeds a threshold score of 0.3681 indicates a likelihood that the individual will be responsive to a DNA-damage therapeutic agent.
  • a cancer is "responsive" to a therapeutic agent if its rate of growth is inhibited as a result of contact with the therapeutic agent, compared to its growth in the absence of contact with the therapeutic agent.
  • Growth of a cancer can be measured in a variety of ways, for instance, the size of a tumor or the expression of tumor markers appropriate for that tumor type may be measured.
  • a cancer is "non-responsive" to a therapeutic agent if its rate of growth is not inhibited, or inhibited to a very low degree, as a result of contact with the therapeutic agent when compared to its growth in the absence of contact with the therapeutic agent.
  • growth of a cancer can be measured in a variety of ways, for instance, the size of a tumor or the expression of tumor markers appropriate for that tumor type may be measured.
  • the quality of being non-responsive to a therapeutic agent is a highly variable one, with different cancers exhibiting different levels of "non-responsiveness" to a given therapeutic agent, under different conditions. Still further, measures of non-responsiveness can be assessed using additional criteria beyond growth size of a tumor, including patient quality of life, degree of metastases, etc.
  • An application of this test will predict end points including, but not limited to, overall survival, progression free survival, radiological response, as defined by RECIST, complete response, partial response, stable disease and serological markers such as, but not limited to, PSA, CEA, CA125, CA15-3 and CA19-9.
  • RNA, DNA or protein within a sample of one or more nucleic acids or their biological derivatives such as encoded proteins may be employed, including quantitative PCR (QPCR), enzyme-linked immunosorbent assay (ELISA) or immunohistochemistry (IHC) and the like.
  • QPCR quantitative PCR
  • ELISA enzyme-linked immunosorbent assay
  • IHC immunohistochemistry
  • the expression profile is compared with a reference or control profile to make a diagnosis regarding the therapy responsive phenotype of the cell or tissue, and therefore host, from which the sample was obtained.
  • the terms "reference” and "control” as used herein in relation to an expression profile mean a standardized pattern of gene or gene product expression or levels of expression of certain biomarkers to be used to interpret the expression classifier of a given patient and assign a prognostic or predictive class.
  • the reference or control expression profile may be a profile that is obtained from a sample known to have the desired phenotype, e.g., responsive phenotype, and therefore may be a positive reference or control profile.
  • the reference profile may be from a sample known to not have the desired phenotype, and therefore be a negative reference profile.
  • this method quantifies the PCR product accumulation through measurement of fluorescence released by a dual-labeled fluorogenic probe (i.e. TaqMan® probe).
  • the obtained expression profile is compared to a single reference profile to obtain information regarding the phenotype of the sample being assayed. In yet other embodiments, the obtained expression profile is compared to two or more different reference profiles to obtain more in depth information regarding the phenotype of the assayed sample. For example, the obtained expression profile may be compared to a positive and negative reference profile to obtain confirmed information regarding whether the sample has the phenotype of interest.
  • the comparison of the obtained expression profile and the one or more reference profiles may be performed using any convenient methodology, where a variety of methodologies are known to those of skill in the array art, e.g., by comparing digital images of the expression profiles, by comparing databases of expression data, etc.
  • Patents describing ways of comparing expression profiles include, but are not limited to, U.S. Pat. Nos. 6,308,170 and 6,228,575, the disclosures of which are herein incorporated by reference. Methods of comparing expression profiles are also described above.
  • the comparison step results in information regarding how similar or dissimilar the obtained expression profile is to the one or more reference profiles, which similarity information is employed to determine the phenotype of the sample being assayed. For example, similarity with a positive control indicates that the assayed sample has a responsive phenotype similar to the responsive reference sample. Likewise, similarity with a negative control indicates that the assayed sample has a non-responsive phenotype to the non-responsive reference sample.
  • the level of expression of a biomarker can be further compared to different reference expression levels.
  • a reference expression level can be a predetermined standard reference level of expression in order to evaluate if expression of a biomarker or biomarker set is informative and make an assessment for determining whether the patient is responsive or non- responsive.
  • determining the level of expression of a biomarker can be compared to an internal reference marker level of expression which is measured at the same time as the biomarker in order to make an assessment for determining whether the patient is responsive or non-responsive.
  • expression of a distinct marker panel which is not comprised of biomarkers of the invention, but which is known to demonstrate a constant expression level can be assessed as an internal reference marker level, and the level of the biomarker expression is determined as compared to the reference.
  • expression of the selected biomarkers in a tissue sample which is a non-tumor sample can be assessed as an internal reference marker level.
  • the level of expression of a biomarker may be determined as having increased expression in certain aspects.
  • the level of expression of a biomarker may be determined as having decreased expression in other aspects.
  • the level of expression may be determined as no informative change in expression as compared to a reference level.
  • the level of expression is determined against a pre-determined standard expression level as determined by the methods provided herein.
  • the invention is also related to guiding conventional treatment of patients.
  • Patients in which the diagnostics test reveals that they are responders to the drugs, of the classes that directly or indirectly affect DNA damage and/or DNA damage repair, can be administered with that therapy and both patient and oncologist can be confident that the patient will benefit.
  • Patients that are designated non-responders by the diagnostic test can be identified for alternative therapies which are more likely to offer benefit to them.
  • the invention further relates to selecting patients for clinical trials where novel drugs of the classes that directly or indirectly affect DNA damage and/or DNA damage repair. Enrichment of trial populations with potential responders will facilitate a more thorough evaluation of that drug under relevant criteria.
  • the invention still further relates to methods of diagnosing patients as having or being susceptible to developing a cancer associated with a DNA damage response deficiency (DDRD).
  • DDRD is defined herein as any condition wherein a cell or cells of the patient have a reduced ability to repair DNA damage, which reduced ability is a causative factor in the development or growth of a tumor.
  • the DDRD diagnosis may be associated with a mutation in the Fanconi anemia/BRCA pathway.
  • the DDRD diagnosis may also be associated with breast cancer or ovarian cancer.
  • These methods of diagnosis comprise the steps of obtaining a test sample from the individual; measuring expression levels of one or more biomarkers in the test sample, wherein the one or more biomarkers are selected from the group consisting of CXCL10, MX1, IDOl, IF144L, CD2, GBP5, PRAME, ITGAL, LRP4, and APOL3; deriving a test score that captures the expression levels; providing a threshold score comprising information correlating the test score and a diagnosis of the cancer; and comparing the test score to the threshold score; wherein the individual is determined to have the cancer or is susceptible to developing the cancer when the test score exceeds the threshold score.
  • One of ordinary skill in the art can determine an appropriate threshold score, and appropriate biomarker weightings, using the teachings provided herein including the teachings of Example 1.
  • the methods of diagnosing patients as having or being susceptible to developing a cancer associated with DDRD comprise measuring expression levels of one or more biomarkers in the test sample, wherein the one or more biomarkers are selected from the group consisting of CXCL10, MX1, IDOl, IF144L, CD2, GBP5, PRAME, ITGAL, LRP4, APOL3, CDR1, FYB, TSPAN7, RAC2, KLHDC7B, GRB14, AC138128.1, KIF26A, CD274, CD 109, ETV7, MFAP5, OLFM4, PI 15, FOSB, FAM19A5, NLRC5, PRICKLE 1, EGR1, CLDN10, ADAMTS4, SP140L, ANXAl, RSAD2, ESR1, IKZF3, OR211P, EGFR, NAT1, LATS2, CYP2B6, PTPRC, PPP1R1A, and AL137218.1.
  • the one or more biomarkers are selected from the group consisting of CX
  • Tables 2A and 2B provide exemplary gene signatures (or gene classifiers) wherein the biomarkers consist of 40 or 44 of the gene products listed therein, respectively, and wherein a threshold score is derived from the individual gene product weightings listed therein.
  • a test score that exceeds a threshold score of 0.3681 indicates a diagnosis of cancer or of being susceptible to developing a cancer.
  • Table 2 The genes determined to be useful in the present methods (Table 2) were identified from gene expression analysis of a cohort of 107 macrodissected breast tumor FFPE tissue samples sourced from the Mayo Clinic Rochester. Ethical approval for this study was obtained from the Institutional Review Board and the Office of Research Ethics Northern Ireland.
  • the Almac's Breast Cancer DSATM research tool has been optimised for analysis of FFPE tissue samples, enabling the use of valuable archived tissue banks.
  • the Almac Breast Cancer DSATM research tool is an innovative microarray platform that represents the transcriptome in both normal and cancerous breast tissues. Consequently, the Breast Cancer DSATM provides a comprehensive representation of the transcriptome within the breast disease and tissue setting, not available using generic microarray platforms. Arrays were scanned using the Affymentrix Genechip® Scanner 7G (Affymetrix Inc., Santa Clara, CA).
  • Quality Control (QC) of profiled samples was carried out using MAS5 pre-processing algorithm. Different technical aspects were addressed: average noise and background homogeneity, percentage of present call (array quality), signal quality, RNA quality and hybridization quality. Distributions and Median Absolute Deviation of corresponding parameters were analyzed and used to identify possible outliers.
  • Almac's Ovarian Cancer DSATM contains probes that primarily target the area within 300 nucleotides from the 3' end of a polynucleotide. Therefore standard Affymetrix RNA quality measures were adapted - for housekeeping genes intensities of 3 ' end probesets along with ratios of 3' end probeset intensity to the average background intensity were used in addition to usual 375' ratios. Hybridization controls were checked to ensure that their intensities and present calls conform to the requirements specified by Affymetrix.
  • mRNA expression level E.avg for each sample was determined by the average expression of all ESR1 probe sets (BRAD.15436_s_at, BRAD.19080_s_at, BREM.1048_at, BRIH.10647Cln2_at, BRIH.5650Cln2_at, BRPD.10690Cln5_at, BRRS.81_at and BRRS.81 -22_at).
  • the mRNA median expression (E. me d.aii) was calculated for all samples. Samples were considered ER positive when E. avg - E. me d.aii> 0.5 and ER negative when E. avg - E. me d.aii ⁇ 0.5.
  • Pre-processing was performed in expression console vl . l with Robust Multi-array Analysis (RMA) (Irizarry et al., 2003) resulting in 2 data matrices of ER positive and ER negative samples composed of 56 and 51 samples respectively.
  • RMA Robust Multi-array Analysis
  • An additional transformation was performed to remove the variance associated with array quality as described by Alter (Alter et al, 2000).
  • a combined background & variance filter was applied to each data matrix to identify the most variable probesets.
  • Hierarchical clustering techniques were applied to microarray data from 199 epithelial serous ovarian tumors analysed using the Ovarian Cancer DSATM (disease specific array) platform (FIG. 1).
  • Raw expression data was preprocessed using the standard Robust Multichip Algorithm (RMA) procedure.
  • RMA Robust Multichip Algorithm
  • 2-D cluster analysis (tumor, probeset) was performed to establish tumor relationships based on the intrinsic list.
  • Hierarchical agglomerative clustering was applied (Pearson correlation distance and Ward's linkage).
  • Optimal partition number was selected using the GAP index (Tibshirani et al., 2002, J. R. Stat. Soc, 63:411-423). All probesets available in the subclusters were mapped to genes names.
  • probesets were mapped to genes (Entrez gene ID) and an enrichment analysis, based on the hypergeometric function (False Discovery Rate applied (Benjamini and Hochberg, 1995, J. R. Stat. Soc. 57:289:300)), was performed. Over-representation of biological processes and pathways were analysed for each gene group generated by the hierarchical clustering for both ER-positive and ER-negative samples using MetacoreTM single experiment analysis workflow from GeneGo®. Antisense probesets were excluded from the analysis. Hypergeometric p-values were assessed for each enriched functional entity class. Functional entity classes with the highest p-values were selected as representative of the group and a general functional category representing these functional entities was assigned to the gene clusters based on significance of representation (i.e. p-value).
  • DDRD DNA-damage response-deficiency
  • the AUC was used to assess the performance of the different models. Iterative Feature Elimination (IFE) was implemented throughout the development of each model, where the maximum AUC was the main criteria in selecting an optimal number of features over cross validation. In cases where there was no visible AUC difference across features, the minimum feature length was selected.
  • IFE Iterative Feature Elimination
  • the expression intensities of the unique genes in the probeset classifier were estimated from the median of the probesets mapping to each gene, excluding anti- sense probesets.
  • a threshold was chosen based on the maximum sensitivity and specificity over all cross validation predictions.
  • the gene level defined expression intensities for the 10 top genes could be used to re-develop the classifier based on only these 10 genes (or any number of features present in current 44 gene signature) by re-estimating classification parameters in cross-validation in the training data set as well as to re-establish the threshold by assessing and maximising the sensitivity and specificity obtained from all cross-validation predictions.
  • the methodology would be similar to the method used when working from a larger feature set (described above) except there will be no feature selection involved: the features will remain the same but will be assigned new weights.
  • the datasets used in for this analysis are namely: FACl [GEO accession number GSE20271, (Tabchy et al, 2010)], FAC2 [GEO accession number GSE22093, (Iwamoto et al, 2011)], FEC [GEO accession number GSE6861, (Bonnefoi et al, 2007)], T/FAC1 [http://bioinformatics.mdanderson.org/pubdata.html, (Hess et al, 2006)], T/FAC2 [GEO accession number GSE16716, (Lee et al, 2010)] and T/FAC3 [GEO accession number GSE20271, (Tabchy et al, 2010)].
  • sample GSM508092 was removed from FACl as it is a metastatic lymph node sample.
  • the objective of this study was to characterize at a transcriptomic level a set of genes that would be capable of determining responsiveness or resistance of a pathogenic cell to DNA- damage therapeutic agents.
  • those samples within the Almac breast cancer dataset that best represented this biology were to be selected and compared to the remaining samples for classifier generation (see next section). It was decided that the samples from sample cluster two within the ER-ve sample set were the most relevant samples for this selection as these showed the greatest proportion of BRCA mutant samples (64%) and they exhibited the most dominant biology (IFN/immune response). From within the ER+ve sample set, the samples from sample cluster two and three were selected as these sample clusters had 73% and 67% BRCA mutant tumors respectively.
  • ER-ve sample set Within the ER-ve sample set, the hierarchical cluster analysis defined three sample clusters and six probeset cluster groups. Probeset cluster three was identified as the most significant biology within the ER-ve sample set and was enriched for interferon and immune response signaling.
  • ER+ve sample set Within the ER+ve sample set, the hierarchical analysis defined six sample groups and six probeset cluster groups. Probeset cluster five was identified as the most significant biology within the ER+ve sample set and was enriched for extracellular matrix remodeling. The next most significant probeset cluster within the ER+ve sample set is probeset cluster six and again was enriched for interferon and immune response signaling.
  • the classification pipeline was used to derive a model using the set of combined ER-ve and ER+ve breast cancer samples.
  • the classification pipeline has been developed in accordance with commonly accepted good practice [MAQC Consortium, Nat Biotechnol 2010].
  • the process will, in parallel: 1) derive gene classification models from empirical data; and 2) assess the classification performance of the models, both under cross-validation.
  • the performance and success of the classifier generation depends on a number of parameters that can be varied, for instance the choice of classification method or probe set filtering.
  • AUC-ROC Receiver Operating Characteristics Curve
  • a cross comparison of the models was made, by first selecting the best number of features for each model based on the highest average AUC, and then using box-plots to visualize the performance for each model. This is demonstrated in FIG. 2. From left to right, the first three plots represent the PLS, SDA and DSDA classifiers respectively that were developed using an initial filtering of probe sets to remove 75% with the lowest average variance and intensity (forcing the inclusion of the gene list). The next three plots respectively represent the PLS, SDA and DSDA classifiers developed using the DDRD (IFN/DNA damage) list only.
  • DDRD IFN/DNA damage
  • Phase II Transformation of the probe set level classification model to a gene level classification model 3.
  • Phase III Validation of re-developed gene classification model using external data sets
  • this model needed to be re-built at the gene level (Phase II). This involved mapping the probe sets in the classification model to the gene level and recalculating the weights for each gene.
  • the 53 probe sets in the selected model mapped to 40 genes listed in Table 2A and subsequently mapped to 44 genes listed in Table 2B when the accuracy of the annotation pipeline was improved through further analysis.
  • the median intensity of all probe sets associated with each gene (Table 2C) is used as the gene expression value. This was calculated for all samples, resulting in a gene expression data matrix, as opposed to a probe set expression data matrix that was used in Phase I for model development and selection. To stabilize the intensities across different batches, the median of all probe sets for each sample was subtracted from the corresponding intensity of each gene for that sample.
  • probe sets that map to the genes in the classifier are determined, excluding anti- sense probe sets (if applicable)
  • the classifier produces a score for each sample, which can then be used to stratify patients from say, more likely to respond to less likely to respond.
  • the performance of the 44-gene DDRD classifier model was validated by the Area Under the ROC (Receiver Operator Characteristic) Curve (AUC) within the original Almac breast dataset and three independent datasets.
  • the AUC is a statistic calculated on the observed disease scale and is a measure of the efficacy of prediction of a phenotype using a classifier model (Wray et. al, PLoS Genetics Vol 6, 1-9).
  • An AUC of 0.5 is typical of a random classifier, and an AUC of 1.0 would represent perfect separation of classes.
  • the 44-gene DDRD classifier model is capable of predicting response to, and selecting patients for, standard breast and ovarian cancer therapeutic drug classes, including DNA damage causing agents and DNA repair targeted therapies, the hypothesis is that the AUCs following application within these datasets should be above 0.5 with the lowest confidence interval also above 0.5.
  • the classifier scores for predicting DDRD status were utilized to assess the ability of the model to separate BRCA mutant samples from sporadic samples. This analysis was performed to assess the relationships between the classifier model and BRCA mutation status. BRCA mutant tumors display a high degree of genomic instability due to a deficiency in DNA damage response by virtue of the loss of functional BRCA 1/2. As such, the hypothesis is that the DDRD classifier models should be able to separate BRCA mutant samples from BRCA wildtype sporadic samples.
  • FIG. 3 shows that the 44-gene classifier models separate the BRCA mutants from the sporadic samples with an AUC of -0.68, where the lower confidence interval is -0.56 for both models (Table 3A); showing that the performance is significantly better than a random classifier.
  • this analysis confirms that the 44-gene DDRD classifier model is capable of identifying samples with high genomic instability due to an inability to repair DNA damage.
  • the third dataset (Bonnefoi et al., Lancet Oncol 8, 1071-1078(2007)) had response data for 66 ER- negative primary breast tumor samples following neoadjuvant 5 -fluorouracil, epirubicin and cyclophosphamide (FEC) treatment.
  • FEC neoadjuvant 5 -fluorouracil, epirubicin and cyclophosphamide
  • Each study used pathological complete response (pCR) or residual disease (RD) as endpoints. As each dataset was relatively small, the data was combined to increase the power of the analysis.
  • Stepwise logistic regression was used to determine the ability of the 44-gene DDRD classifier model to predict response in the combined datasets when adjusting for clinical variables (Table 4).
  • the 44-gene DDRD classifier model was determined to be the most significant clinical variable in univariate analysis. Multivariate analysis confirmed that the 44- gene DDRD classifier model's predictive value was independent of stage, grade and notably ER status.
  • Negativity for estrogen, progesterone and HER2 receptors has been suggested as a biomarker of abnormal DDR and thus response to DNA-damaging and DNA repair targeted therapies (Foulkes et al, 2010).
  • this approach excludes the 20% of BRCA1 and the 40% of BRCA2 mutant tumors that are reported to be ER-positive (Foulkes et al., 2004; Tung et al., 2010).
  • the 44-gene DDRD classifier detects the DDRD subgroup in both ER-positive and ER-negative tumors, as validated by the multivariate analysis of the 44-gene DDRD classifier's predictive value within the combined analysis of FEC and FAC datasets, demonstrating its independence from ER status. Clinically, this is an important aspect of the translational application of the DDRD classifier as it suggests it can be applied to all breast cancer patients, irrespective of ER status, to determine their predicted responsiveness to DNA-damaging therapeutics. (2) Assessment of 44-gene DDRD classifier model's predictive power to taxane-containing chemotherapy regimens
  • the Fanconi anemia/BRCA (FA/BRCA) pathway which includes BRCA1 and BRCA2, plays an integral role in DNA repair and can be lost in breast cancer either due to mutation or epigenetic silencing (Kennedy and DAndrea, 2006). It was therefore determined if the 44-gene DDRD classifier model could detect abrogation of members of this pathway in addition to BRCA1 and BRCA2.
  • a public dataset was identified with microarray data generated from the bone marrow of 21 FA patients carrying a range of mutations in the FA/BRCA pathway and 11 healthy controls with a functional FA/BRCA pathway (Vanderwerf, S. M., et al., Blood 114, 5290-5298 (2009).
  • the 44-gene DDRD classifier model is able to significantly separate BRCA mutant breast tumor samples from wildtype BRCA (sporadic) breast tumor samples. This implies that the DDRD classifier model is capable of detecting biology related to tumors with a high level of genomic instability, such as BRCA mutant tumors. These tumors typically respond better to DNA damaging chemotherapeutic regimens.
  • the 44-gene DDRD classifier model is able to significantly separate defined responders (those that demonstrated pCR) from the non-responders (those that did not demonstrate pCR) in a combination of three independent breast datasets following neoadjuvant treatment with FAC and FEC (Bonnefoi et al., 2007; Iwamoto et al., 2011; Tabchy et al.,
  • the 44-gene DDRD classifier model is able to significantly separate responders from non-responders within an independent Almac ovarian dataset following adjuvant treatment with platinum or platinum/taxane based therapy. This data was generated using FFPE samples profiled upon the Almac Ovarian DSATM.
  • the 44-gene DDRD classifier model is able to significantly distinguish between FA/BRCA mutant and normal samples using bone marrow tissue samples, demonstrating a strong correlation between the DDRD classifier and dysfunction of the FA/BRCA pathway through multiple mechanisms.
  • the DDRD classifier model has been independently validated and demonstrated robustness in performance across three different disease areas (breast, ovarian and FA), demonstrated ability to separate responders from non-responders to four different chemotherapeutic regimens (FAC, FEC, T/FAC and platinum/taxane) in two different sample types (FFPE and FF) utilizing data from four different microarray platforms (Almac Breast DSATM and Almac Ovarian DSATM, Affymetrix X3P microarray and Affymetrix U133A microarray). It has been demonstrated that the DDRD is an independent predictor of response to DNA-damage therapeutic agents and can predict mutations in the FA/BRCA pathways.
  • This plasticity and repeatability of performance implies that the biology identified within the DDRD subgroup identified via the 44-gene classifier model is significantly and robustly related to predicting response to DNA damage causing agents and as such supports the claim of this invention which is to identify a subtype that can be used to predict response to, and select patients for, standard breast and ovarian cancer therapeutic drug classes, including drugs that damage DNA directly, damage DNA indirectly or inhibit normal DNA damage signaling and/or repair processes.
  • AUC Area Under the Receiver Operating Characteristics Curve
  • ACC Accuracy
  • SENS Sensitivity
  • SPEC Specificity
  • PPV Positive Predictive value
  • NPV Negative Predictive Value
  • RR Relative Risk
  • pCR pathological complete response
  • RD residual disease.
  • HCC1937 parental, HCC1937-EV and HCC1937-BR cell-lines were kindly donated by Professor Paul Harkin from Queen's University College Birmingham (QUB).
  • the cell-lines were routinely maintained in RPMI-1640 medium supplemented with 50 U penicillin/ml, 50 g streptomycin/ml, 2mM glutamine, ImM Sodium Pyruvate and 20% (v/v) fetal bovine serum (FBS).
  • FBS v/v fetal bovine serum
  • the HCC1937-EV and HCC937-BR cell-lines also required 0.2ml/mg geneticin.
  • Cell- lines were cultured at 37°C with a humidified atmosphere of 5% C0 2 .
  • the DDRD subgroup can be identified within breast cancer cell-line models
  • HCC1937 breast cancer cell-line is DDRD due to a BRCAl mutation (Tomlinson et al., 1998).
  • the 44-gene classifier was applied to HCC1937 empty vector control cells (HCC1937-EV) and HCC1937 cells in which BRCAl functionality was corrected (HCC1937-BR) (FIG. 7A).
  • HCC1937-EV empty vector control cells
  • HCC1937-BR HCC1937-BR
  • the DDRD 44-gene classifier score was found to be higher within HCC1937-EV relative to HCC1937-BR cells, with average scores of 0.5111 and 0.1516 respectively (FIG. 7B).
  • the HCC1937 BRCAl mutant cell-line was more sensitive to the PARP-1 inhibitor KU0058948 (FIG. 7C) and cisplatin (FIG. 7D) relative to the BRCAl corrected cell-line.
  • the DDRD 44-gene classifier measures immune signalling in DDRD-positive tumor cells and correlates with response to both a DNA-damaging agent (cisplatin) and a DNA repair targeted agent (PARP-1 inhibitor).
  • the DDRD 44-gene classifier detects dysfunction of the Fanconi Anemia/BRCA pathway
  • the Fanconi anemia/BRCA (FA/BRCA) pathway which includes BRCAl and BRCA2, plays an integral role in DNA repair and can be lost in breast cancer either due to mutation or epigenetic silencing (Kennedy, R. D., and DAndrea, A. D., J Clin Oncol 24, 3799-3808 (2006)). It was determined if the DDRD 44-gene classifier could detect abrogation of members of this pathway in addition to BRCAl and BRCA2.
  • a public dataset was identified with microarray data generated from the bone marrow of 21 FA patients carrying a range of mutations in the FA/BRCA pathway and 11 healthy controls with a functional FA/BRCA pathway (Vanderwerf et al., 2009).
  • the DDRD 44-gene classifier score was significantly higher in the BRCA1 mutant, and thus DDRD, HCC1937 breast cancer cell-line relative to an isogenic BRCA1 corrected cell-line. As the 44-gene classifier score correlates with DDR dysfunction within these cells, it demonstrates that the immune signalling detected by the DDRD classifier is intrinsic to the cell and not a function of lymphocytic infiltrate.
  • BRCA1 and BRCA2 represent part of the FA/BRCA DDR network, which contains a number of other proteins that have been reported to be mutant or under-expressed in approximately 33% of breast cancer (Kennedy, R. D., and DAndrea, A.
  • the DDRD 44- gene classifier significantly separated bone marrow samples from patients with FA mutations from normal controls. This suggests that the DDRD classifier is capable of detecting any abnormality within the pathway rather than specifically BRCA1 or BRCA2 dysfunction. It is possible that the DDRD 44-gene classifier may identify tumors with DDR-deficiency due to other mechanisms such as PTEN loss, cell-cycle checkpoint dysfunction or increased reactive oxygen species due to metabolic disturbance. Due to constitutive DNA-damage, these tumors are likely to respond to DNA repair targeted therapies such as PARP-1 or CHKl/2 inhibitors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Medical Informatics (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Public Health (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Evolutionary Biology (AREA)
  • Theoretical Computer Science (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Epidemiology (AREA)
  • Data Mining & Analysis (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Biomedical Technology (AREA)
  • Databases & Information Systems (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • General Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Evolutionary Computation (AREA)
  • Software Systems (AREA)
  • Artificial Intelligence (AREA)
  • Bioethics (AREA)

Abstract

Cette invention concerne des méthodes et des compositions d'identification d'un test de diagnostic moléculaire du cancer. Ce test définit un nouveau sous-type moléculaire déficient en réparation des lésions de l'ADN et permet la classification d'un patient dans ce sous-type. Cette invention peut être utilisée pour déterminer si des patients atteints d'un cancer sont cliniquement sensibles ou insensibles à un schéma thérapeutique avant administration de toute chimiothérapie. Ce test peut être utilisé dans différents types de cancer et avec différents médicaments qui affectent directement ou indirectement les lésions ou les réparations de l'ADN, comme bon nombre des agents chimiothérapeutiques cytotoxiques standard actuellement utilisés. L'invention concerne en particulier l'utilisation de certaines associations de marqueurs prédictifs, l'expression desdits marqueurs prédictifs étant corrélée avec la sensibilité ou l'insensibilité du patient à un schéma thérapeutique.
EP11825959.7A 2010-09-15 2011-09-15 Aissaies moleculaires pour predire de la reponse d'un patient atteint d'un cancer a un agent therapeutique del'adn endommage Active EP2619574B1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP15181181.7A EP2975399B1 (fr) 2010-09-15 2011-09-15 Test de diagnostic moléculaire pour le cancer

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US38320110P 2010-09-15 2010-09-15
US201161490039P 2011-05-25 2011-05-25
PCT/US2011/051803 WO2012037378A2 (fr) 2010-09-15 2011-09-15 Test de diagnostic moléculaire du cancer

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP15181181.7A Division-Into EP2975399B1 (fr) 2010-09-15 2011-09-15 Test de diagnostic moléculaire pour le cancer
EP15181181.7A Division EP2975399B1 (fr) 2010-09-15 2011-09-15 Test de diagnostic moléculaire pour le cancer

Publications (3)

Publication Number Publication Date
EP2619574A2 true EP2619574A2 (fr) 2013-07-31
EP2619574A4 EP2619574A4 (fr) 2014-03-12
EP2619574B1 EP2619574B1 (fr) 2020-10-28

Family

ID=45832248

Family Applications (2)

Application Number Title Priority Date Filing Date
EP11825959.7A Active EP2619574B1 (fr) 2010-09-15 2011-09-15 Aissaies moleculaires pour predire de la reponse d'un patient atteint d'un cancer a un agent therapeutique del'adn endommage
EP15181181.7A Active EP2975399B1 (fr) 2010-09-15 2011-09-15 Test de diagnostic moléculaire pour le cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP15181181.7A Active EP2975399B1 (fr) 2010-09-15 2011-09-15 Test de diagnostic moléculaire pour le cancer

Country Status (16)

Country Link
US (4) US9670547B2 (fr)
EP (2) EP2619574B1 (fr)
JP (2) JP2013537045A (fr)
KR (1) KR20130115250A (fr)
CN (2) CN103299188B (fr)
AU (2) AU2011302004B2 (fr)
BR (1) BR112013006207A2 (fr)
CA (1) CA2811015A1 (fr)
EA (1) EA201390370A1 (fr)
HK (1) HK1216665A1 (fr)
MX (1) MX344636B (fr)
MY (1) MY166040A (fr)
NZ (3) NZ712823A (fr)
SG (2) SG10201610027VA (fr)
WO (1) WO2012037378A2 (fr)
ZA (2) ZA201301567B (fr)

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5645816B2 (ja) 2009-05-25 2014-12-24 国立大学法人東京工業大学 中枢神経細胞の増殖及び分化に係る中核因子を含む医薬組成物
AU2011302004B2 (en) 2010-09-15 2015-10-29 Almac Diagnostics Limited Molecular diagnostic test for cancer
WO2012149299A2 (fr) 2011-04-29 2012-11-01 Celgene Corporaiton Procédés de traitement du cancer et de maladies inflammatoires au moyen de céréblon en tant que prédicteur
EP2705370A2 (fr) * 2011-05-06 2014-03-12 Xentech Marqueurs pour le pronostic et la thérapie du cancer, et procédés d'utilisation
EA201691257A1 (ru) 2011-06-02 2017-02-28 Олмак Дайэгностикс Лимитед Молекулярно-диагностический тест на рак
EP2723898A4 (fr) * 2011-06-22 2015-09-30 Oncocyte Corp Méthodes et compositions pour traitement et diagnostique d'un cancer de la vessie
EP3904875A1 (fr) * 2012-06-29 2021-11-03 Celgene Corporation Procédés pour déterminer l'efficacité d'un médicament en utilisant ikzf3 (aiolos)
US9587281B2 (en) 2012-08-14 2017-03-07 Celgene Corporation Cereblon isoforms and their use as biomarkers for therapeutic treatment
BR112015005243A2 (pt) * 2012-09-10 2017-07-04 Celgene Corp métodos para o tratamento de câncer de mama localmente avançado
US11091809B2 (en) 2012-12-03 2021-08-17 Almac Diagnostic Services Limited Molecular diagnostic test for cancer
US9302005B2 (en) 2013-03-14 2016-04-05 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
CA2905798C (fr) * 2013-03-15 2023-01-24 Genentech, Inc. Biomarqueurs et methodes de traitement d'etats associes a pd-1 et pd-l1
HUE052930T2 (hu) * 2013-04-09 2021-05-28 Univ Illinois DNQ vagy DNQ-87 alkalmazása PARP1-inhibitorral kombinációban rák kezelésére
GB201316024D0 (en) * 2013-09-09 2013-10-23 Almac Diagnostics Ltd Molecular diagnostic test for lung cancer
GB201316027D0 (en) * 2013-09-09 2013-10-23 Almac Diagnostics Ltd Molecular diagnostic test for oesophageal cancer
EP3052131B1 (fr) 2013-10-01 2018-12-05 Mayo Foundation for Medical Education and Research Procédés de traitement du cancer chez des patients présentant des taux élevés de bim
AU2015203904B2 (en) * 2014-01-06 2018-07-19 Expression Pathology, Inc. SRM assay for PD-L1
GB201409479D0 (en) 2014-05-28 2014-07-09 Almac Diagnostics Ltd Molecular diagnostic test for cancer
EP3137076A4 (fr) * 2014-05-02 2017-12-06 Emory University Traitements sélectifs de chimiothérapie et méthodes de diagnostic associées à ceux-ci
WO2015179654A1 (fr) 2014-05-22 2015-11-26 Mayo Foundation For Medical Education And Research Distinction d'anticorps anti-b7-h1 agonistes et antagonistes
WO2015200795A1 (fr) 2014-06-27 2015-12-30 Celgene Corporation Compositions et procédés pour induire des modifications conformationnelles dans céréblon et d'autres ubiquitine ligases e3
CN106604933B (zh) 2014-07-11 2021-08-10 文塔纳医疗系统公司 抗pd-l1抗体及其诊断用途
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
GB2553736A (en) 2015-05-19 2018-03-14 S Sridhar T Method for identification of a deficient BRCA1 function
GB201512869D0 (en) * 2015-07-21 2015-09-02 Almac Diagnostics Ltd Gene signature for minute therapies
ES2742197T7 (es) 2015-07-23 2021-10-06 Inst Curie Uso de una combinación de molécula Dbait e inhibidores de PARP para tratamiento del cáncer
JP6624704B2 (ja) 2015-08-31 2019-12-25 日立化成株式会社 尿路上皮疾患の評価のための分子法
CN108474040B (zh) * 2015-10-09 2023-05-16 夸登特健康公司 使用无细胞dna的基于群体的治疗推荐
WO2017075045A2 (fr) 2015-10-30 2017-05-04 Mayo Foundation For Medical Education And Research Anticorps anti-b7-h1
US10830762B2 (en) 2015-12-28 2020-11-10 Celgene Corporation Compositions and methods for inducing conformational changes in cereblon and other E3 ubiquitin ligases
US20170321285A1 (en) * 2016-05-03 2017-11-09 The Texas A&M University System Nlrc5 as a biomarker for cancer patients and a target for cancer therapy
WO2017216559A1 (fr) * 2016-06-14 2017-12-21 Almac Diagnostics Limited Prédiction de la sensibilité à une thérapie dans le cancer de la prostate
JP7045724B2 (ja) 2016-11-07 2022-04-01 ニューラクル サイエンス カンパニー リミテッド 配列類似性を持つ抗ファミリー19、メンバーa5抗体及びその用途
CN108663519A (zh) * 2017-04-21 2018-10-16 江苏希摩生物科技有限公司 快速预测和提高BRCA1/2野生型卵巢癌细胞对Olaparib敏感性的方法
WO2018213141A1 (fr) * 2017-05-16 2018-11-22 Hitachi Chemical Co. America, Ltd. Procédés de détection du cancer de l'ovaire à l'aide de vésicules extracellulaires pour analyse moléculaire
KR102340352B1 (ko) * 2017-06-27 2021-12-21 주식회사 뉴라클사이언스 섬유증의 치료를 위한 항-fam19a5 항체의 용도
US11746149B2 (en) 2017-06-27 2023-09-05 Neuracle Science Co., Ltd. Use of anti-family with sequence similarity 19, member A5 antibodies for the treatment of glaucoma
CN111093700A (zh) * 2017-06-27 2020-05-01 纽洛可科学有限公司 抗fam19a5抗体用于治疗癌症的用途
AU2018291082A1 (en) 2017-06-27 2020-01-16 Neuracle Science Co., Ltd. Anti-FAM19A5 antibodies and uses thereof
CN110021357B (zh) * 2017-12-27 2021-06-04 浙江安诺优达生物科技有限公司 模拟癌症基因组测序数据生成装置
FR3078343A1 (fr) 2018-02-27 2019-08-30 Lxrepair Methode pour generer un profil des capacites de reparation de l'adn de cellules tumorales et ses applications
CN108379584B (zh) * 2018-04-12 2020-08-11 上海交复生物医药科技有限公司 RSAD2通过wnt通路影响肿瘤细胞替莫唑胺耐药性
JP7171081B2 (ja) 2018-04-24 2022-11-15 ニューラクル サイエンス カンパニー リミテッド 神経病性疼痛の治療のための配列類似性19、メンバーa5抗体を有する抗ファミリーの用途
CN108977547B (zh) * 2018-08-14 2019-05-28 江门市中心医院 Tspan1在乳腺癌转移的诊断、预后及治疗中的应用
CN109295208A (zh) * 2018-10-26 2019-02-01 德阳市人民医院 Pi15作为骨关节炎标志物的应用
CN109652540A (zh) * 2018-12-21 2019-04-19 思泰得精准(北京)医学检验实验室有限公司 一种甲状腺癌早期检测分子标志物及其应用
WO2020136603A1 (fr) * 2018-12-27 2020-07-02 Neuracle Science Co., Ltd. Utilisation d'anticorps anti-fam19a5 pour le traitement de l'athérosclérose
WO2020172671A1 (fr) * 2019-02-22 2020-08-27 Advantagene, Inc. Polythérapie avec du gmci et du ddri pour le traitement du cancer
CN110570922B (zh) * 2019-07-19 2022-06-10 浙江大学 一种评估hr缺陷模型及应用
CA3156171A1 (fr) * 2019-09-30 2021-04-08 Candel Therapeutics, Inc. Traitement anticancereux par immunotherapie cytotoxique a mediation genique et inhibiteur atr combines
WO2023017525A1 (fr) * 2021-08-11 2023-02-16 OncoHost Ltd. Prédiction de la réponse d'un patient

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008005281A2 (fr) * 2006-06-30 2008-01-10 Rosetta Inpharmatics Llc Gènes associés à une réponse à une chimiothérapie et leurs utilisations

Family Cites Families (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5525464A (en) 1987-04-01 1996-06-11 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
US5202231A (en) 1987-04-01 1993-04-13 Drmanac Radoje T Method of sequencing of genomes by hybridization of oligonucleotide probes
GB8810400D0 (en) 1988-05-03 1988-06-08 Southern E Analysing polynucleotide sequences
US5547839A (en) 1989-06-07 1996-08-20 Affymax Technologies N.V. Sequencing of surface immobilized polymers utilizing microflourescence detection
US5143854A (en) 1989-06-07 1992-09-01 Affymax Technologies N.V. Large scale photolithographic solid phase synthesis of polypeptides and receptor binding screening thereof
US5800992A (en) 1989-06-07 1998-09-01 Fodor; Stephen P.A. Method of detecting nucleic acids
US6040138A (en) 1995-09-15 2000-03-21 Affymetrix, Inc. Expression monitoring by hybridization to high density oligonucleotide arrays
EP0430881A3 (en) 1989-11-29 1991-10-23 Ciba-Geigy Ag Photochromic compounds, process for their preparation and their use
US5288644A (en) 1990-04-04 1994-02-22 The Rockefeller University Instrument and method for the sequencing of genome
US5324633A (en) 1991-11-22 1994-06-28 Affymax Technologies N.V. Method and apparatus for measuring binding affinity
EP0636186B1 (fr) 1992-04-03 1998-11-25 The Perkin-Elmer Corporation Procede et composition de sondage
US5503980A (en) 1992-11-06 1996-04-02 Trustees Of Boston University Positional sequencing by hybridization
US5858659A (en) 1995-11-29 1999-01-12 Affymetrix, Inc. Polymorphism detection
US5470710A (en) 1993-10-22 1995-11-28 University Of Utah Automated hybridization/imaging device for fluorescent multiplex DNA sequencing
GB9401833D0 (en) 1994-02-01 1994-03-30 Isis Innovation Method for discovering ligands
WO1997027317A1 (fr) 1996-01-23 1997-07-31 Affymetrix, Inc. Evaluation rapide de difference d'abondance d'acides nucleiques, avec un systeme d'oligonucleotides haute densite
GB9507238D0 (en) 1995-04-07 1995-05-31 Isis Innovation Detecting dna sequence variations
US5661028A (en) 1995-09-29 1997-08-26 Lockheed Martin Energy Systems, Inc. Large scale DNA microsequencing device
AU2189397A (en) 1996-02-08 1997-08-28 Affymetrix, Inc. Chip-based speciation and phenotypic characterization of microorganisms
EP1002264B1 (fr) 1997-07-25 2004-04-14 Affymetrix, Inc. (a Delaware Corporation) Procede d'obtention d'une base de donnees bioinformatique
US7324926B2 (en) * 1999-04-09 2008-01-29 Whitehead Institute For Biomedical Research Methods for predicting chemosensitivity or chemoresistance
US20040146921A1 (en) 2003-01-24 2004-07-29 Bayer Pharmaceuticals Corporation Expression profiles for colon cancer and methods of use
US7601505B2 (en) 2003-05-29 2009-10-13 Millennium Pharmaceuticals, Inc. Compositions, kits, and methods for identification, assessment, prevention, and therapy of breast cancer
BRPI0414446A (pt) 2003-09-18 2006-11-14 Genmab As métodos para triar um agente terapêutico, para a supressão da sequência de polinucleotìdeo, para o tratamento de um cáncer não dependente de esteróide, para triar a ligação de um agente especificamente a um polinucleotìdeo e para determinar se um paciente está em risco de desenvolver ou ter um cáncer não dependente de esteróide, composição farmacêutica, usos de um agente terapêutico, de uma molécula anti-sentido ou de uma célula que expressa e/ou contém a molécula anti-sentido, de pelo menos uma das proteìnas de membrana imunogênicas, fragmentos, derivados ou homólogos destas ou de uma célula que contém e/ou que expressa pelo menos uma das proteìnas de membranas imunogênicas ou fragmentos, derivados ou homólogos destas e de um agente ou anticorpo, agente, e, kit para identificar um paciente em risco de desenvolver ou ter cáncer não dependente de esteróide
CA2547077C (fr) * 2003-12-01 2015-11-03 Kudos Pharmaceuticals Limited Inhibiteurs de reparation d'adn endommage pour le traitement du cancer
US20050287544A1 (en) 2003-12-01 2005-12-29 Francois Bertucci Gene expression profiling of colon cancer with DNA arrays
CN1905864B (zh) * 2003-12-01 2011-04-06 库多斯药物有限公司 用于治疗癌症的dna损伤修复抑制剂
WO2005083440A2 (fr) 2004-02-19 2005-09-09 Yale University Identification de biomarqueurs proteiques du cancer par des techniques proteomiques
WO2005083128A2 (fr) 2004-02-25 2005-09-09 University Of South Florida Methodes de prevision de l'issue d'un cancer et signatures genetiques utilisees dans celles-ci
US20060195266A1 (en) 2005-02-25 2006-08-31 Yeatman Timothy J Methods for predicting cancer outcome and gene signatures for use therein
AU2005300688B2 (en) 2004-11-03 2012-02-02 Almac Diagnostics Limited Transcriptome microarray technology and methods of using the same
CA2624086A1 (fr) 2005-09-28 2007-04-05 H. Lee Moffitt Cancer Center Traitements anticancereux individualises
EP1777523A1 (fr) 2005-10-19 2007-04-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthode pour pronostiquer la progression d'un cancer et les résultats du patient et les moyens pour performer la méthode
US8445198B2 (en) 2005-12-01 2013-05-21 Medical Prognosis Institute Methods, kits and devices for identifying biomarkers of treatment response and use thereof to predict treatment efficacy
BRPI0706511A2 (pt) 2006-01-11 2011-03-29 Genomic Health Inc marcadores de expressão genética para prognóstico de cáncer colorretal
US7892740B2 (en) * 2006-01-19 2011-02-22 The University Of Chicago Prognosis and therapy predictive markers and methods of use
US20070218512A1 (en) 2006-02-28 2007-09-20 Alex Strongin Methods related to mmp26 status as a diagnostic and prognostic tool in cancer management
WO2007112330A2 (fr) 2006-03-24 2007-10-04 Diadexus, Inc. Compositions et méthodes pour détecter, pronostiquer et traiter un cancer du côlon
US8768629B2 (en) 2009-02-11 2014-07-01 Caris Mpi, Inc. Molecular profiling of tumors
EP2390363A1 (fr) * 2006-06-02 2011-11-30 GlaxoSmithKline Biologicals s.a. Procédé d'identification si un patient est réactif ou non à l'immunothérapie basé sur l'expression différentielle du gène PRF1
AU2007262635B2 (en) * 2006-06-23 2014-09-11 Adc Therapeutics Sa Polynucleotides and polypeptide sequences involved in cancer
WO2008066624A2 (fr) * 2006-10-20 2008-06-05 Dana-Farber Cancer Institute Compositions et procédés pour traiter le cancer
WO2008089465A2 (fr) 2007-01-18 2008-07-24 University Of Southern California Polymorphismes géniques comme prédicteurs spécifiques au sexe dans la thérapie contre le cancer
EP1961825A1 (fr) 2007-02-26 2008-08-27 INSERM (Institut National de la Santé et de la Recherche Medicale) Procédé pour prévoir l'apparition de métastase dans les patients souffrant d'un cancer du sein
WO2008132176A2 (fr) 2007-04-27 2008-11-06 Universite Catholique De Louvain Méthode de prévision de la réponse d'un patient à une thérapie bloquant le tnf
CA2730277A1 (fr) 2008-07-08 2010-01-14 Source Precision Medicine, Inc. D/B/A Source Mdx Profilage de l'expression genique pour la prediction de la survie de sujets atteints d'un cancer de la prostate
WO2010040083A2 (fr) 2008-10-03 2010-04-08 The U.S.A., As Represented By The Secretary, Department Of Health And Human Services Prédicteurs de chimiorésistance par expression génique
US20110295782A1 (en) 2008-10-15 2011-12-01 Alexander Stojadinovic Clinical Decision Model
WO2011109440A1 (fr) 2010-03-01 2011-09-09 Caris Life Sciences Luxembourg Holdings Biomarqueurs pour théranostique
WO2010060055A1 (fr) 2008-11-21 2010-05-27 Duke University Prédiction du risque et du succès d’un traitement du cancer
WO2010118782A1 (fr) * 2009-04-17 2010-10-21 Universite Libre De Bruxelles Procédés et outils pour prédire l'efficacité d'anthracyclines dans le traitement du cancer
WO2011005384A2 (fr) * 2009-05-29 2011-01-13 Baylor College Of Medicine Réparation de l'adn ou signature de gène de type brca1
WO2011153545A2 (fr) 2010-06-04 2011-12-08 Bioarray Therapeutics, Inc. Signature d'expression génique utilisée pour prédire une réponse à une chimiothérapie lors d'un cancer du sein
AU2011302004B2 (en) 2010-09-15 2015-10-29 Almac Diagnostics Limited Molecular diagnostic test for cancer

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008005281A2 (fr) * 2006-06-30 2008-01-10 Rosetta Inpharmatics Llc Gènes associés à une réponse à une chimiothérapie et leurs utilisations

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
AKIO OOYAMA ET AL: "Gene expression analysis using human cancer xenografts to identify novel predictive marker genes for the efficacy of 5-fluorouracil-based drugs", CANCER SCIENCE, vol. 97, no. 6, 1 June 2006 (2006-06-01), pages 510-522, XP55048094, ISSN: 1347-9032, DOI: 10.1111/j.1349-7006.2006.00204.x *
FARMER P ET AL: "A stroma-related gene signature predicts resistance to epirubicin-containing neoadjuvant chemotherapy in breast cancer", BREAST CANCER RESEARCH AND TREATMENT, SPRINGER, NEW YORK, NY, vol. 106, no. Suppl. 1, 3 November 2007 (2007-11-03), page S11, XP002531722, ISSN: 0167-6806 *
KORRAT ANDRE ET AL: "Gene signature-based prediction of tumor response to cyclophosphamide", CANCER GENOMICS & PROTEOMICS, INTERNATIONAL INSTITUTE OF ANTICANCER RESEARCH, GR, vol. 4, no. 3, 1 May 2007 (2007-05-01), pages 187-195, XP009175760, ISSN: 1109-6535 *
RIKO KAWANO ET AL: "Oncogene Associated cDNA Microarray Analysis Shows PRAME Gene Expression is a Marker for Response to Anthracycline Containing Chemotherapy in Patients with Diffuse Large B-cell Lymphoma", JOURNAL OF CLINICAL AND EXPERIMENTAL HEMATOPATHOLOGY, vol. 49, no. 1, 1 May 2009 (2009-05-01), pages 1-7, XP055098523, ISSN: 1346-4280, DOI: 10.3960/jslrt.49.1 *
RODRIGUEZ A A ET AL: "DNA repair signature is associated with anthracycline response in triple negative breast cancer patients", BREAST CANCER RESEARCH AND TREATMENT, KLUWER ACADEMIC PUBLISHERS, BO, vol. 123, no. 1, 26 June 2010 (2010-06-26) , pages 189-196, XP019814685, ISSN: 1573-7217 *
SUSAN CLEATOR ET AL: "Gene expression patterns for doxorubicin (Adriamycin) and cyclophosphamide (Cytoxan) (AC) response and resistance", BREAST CANCER RESEARCH AND TREATMENT, KLUWER ACADEMIC PUBLISHERS, BO, vol. 95, no. 3, 1 February 2006 (2006-02-01), pages 229-233, XP019274969, ISSN: 1573-7217 *
TSAO D A ET AL: "Gene Expression Profiles for Predicting the Efficacy of the Anticancer Drug 5-Fluorouracil in Breast Cancer", DNA AND CELL BIOLOGY, MARY ANN LIEBERT, NEW YORK, NY, US, vol. 29, no. 6, 17 June 2010 (2010-06-17), pages 285-293, XP002712321, ISSN: 1044-5498, DOI: 10.1089/DNA.2009.1006 [retrieved on 2010-05-18] *

Also Published As

Publication number Publication date
AU2016200494B2 (en) 2017-11-16
MY166040A (en) 2018-05-21
ZA201301567B (en) 2016-04-28
EA201390370A1 (ru) 2013-07-30
CN103299188B (zh) 2017-03-15
WO2012037378A2 (fr) 2012-03-22
ZA201508689B (en) 2021-05-26
WO2012037378A3 (fr) 2012-07-05
KR20130115250A (ko) 2013-10-21
US20170198360A1 (en) 2017-07-13
US9677140B2 (en) 2017-06-13
JP2013537045A (ja) 2013-09-30
US20140031260A1 (en) 2014-01-30
EP2619574B1 (fr) 2020-10-28
NZ712823A (en) 2017-05-26
AU2011302004B2 (en) 2015-10-29
CN103299188A (zh) 2013-09-11
AU2016200494A1 (en) 2016-04-07
US20160060705A1 (en) 2016-03-03
EP2975399A1 (fr) 2016-01-20
MX2013002879A (es) 2013-05-17
BR112013006207A2 (pt) 2017-10-24
NZ620799A (en) 2015-10-30
SG10201610027VA (en) 2017-01-27
US9670547B2 (en) 2017-06-06
CN106978480A (zh) 2017-07-25
US10214777B2 (en) 2019-02-26
JP2017093440A (ja) 2017-06-01
SG188397A1 (en) 2013-04-30
EP2975399B1 (fr) 2022-05-11
US20140051591A1 (en) 2014-02-20
MX344636B (es) 2017-01-04
AU2011302004A1 (en) 2013-04-04
US10378066B2 (en) 2019-08-13
EP2619574A4 (fr) 2014-03-12
CA2811015A1 (fr) 2012-03-22
HK1216665A1 (zh) 2016-11-25
NZ608459A (en) 2015-10-30

Similar Documents

Publication Publication Date Title
US10378066B2 (en) Molecular diagnostic test for cancer
EP3325653B1 (fr) Signature de genes pour thérapies immunitaires pour le cancer
US10260097B2 (en) Method of using a gene expression profile to determine cancer responsiveness to an anti-angiogenic agent
US20160222459A1 (en) Molecular diagnostic test for lung cancer
US20160002732A1 (en) Molecular diagnostic test for cancer
AU2012261820A1 (en) Molecular diagnostic test for cancer
US20160222460A1 (en) Molecular diagnostic test for oesophageal cancer
WO2017216559A1 (fr) Prédiction de la sensibilité à une thérapie dans le cancer de la prostate

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130410

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

RIN1 Information on inventor provided before grant (corrected)

Inventor name: HILL, LAURA A.

Inventor name: DAVISON, TIMOTHY

Inventor name: KENNEDY, RICHARD

Inventor name: MCDYER, FIONNUALA

Inventor name: GOFFARD, NICOLAS

Inventor name: DEHARO, STEVE

Inventor name: PROUTSKI, VITALI

Inventor name: BYLESJO, MAX

Inventor name: HARKIN, DENIS PAUL

Inventor name: KEATING, KATHERINE E.

Inventor name: O'DONNELL, JUDE

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ALMAC DIAGNOSTICS LIMITED

RIN1 Information on inventor provided before grant (corrected)

Inventor name: PATTERSON, FIONNUALA

Inventor name: KEATING, KATHERINE E.

Inventor name: GOFFARD, NICOLAS

Inventor name: DAVISON, TIMOTHY

Inventor name: HARKIN, DENIS PAUL

Inventor name: HILL, LAURA A.

Inventor name: BYLESJO, MAX

Inventor name: KENNEDY, RICHARD

Inventor name: PROUTSKI, VITALI

Inventor name: DEHARO, STEVE

Inventor name: O'DONNELL, JUDE

A4 Supplementary search report drawn up and despatched

Effective date: 20140207

RIC1 Information provided on ipc code assigned before grant

Ipc: G01N 33/48 20060101AFI20140203BHEP

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1186771

Country of ref document: HK

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20170214

RIC1 Information provided on ipc code assigned before grant

Ipc: C12Q 1/6886 20180101AFI20180628BHEP

RIC1 Information provided on ipc code assigned before grant

Ipc: C12Q 1/6886 20180101AFI20180628BHEP

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: ALMAC DIAGNOSTIC SERVICES LIMITED

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

RIN1 Information on inventor provided before grant (corrected)

Inventor name: DAVISON, TIMOTHY

Inventor name: O'DONNELL, JUDE

Inventor name: PATTERSON, FIONNUALA

Inventor name: PROUTSKI, VITALI

Inventor name: KEATING, KATHERINE E.

Inventor name: DEHARO, STEVE

Inventor name: HARKIN, DENIS PAUL

Inventor name: HILL, LAURA A.

Inventor name: GOFFARD, NICOLAS

Inventor name: KENNEDY, RICHARD

Inventor name: BYLESJO, MAX

INTG Intention to grant announced

Effective date: 20200127

RIN1 Information on inventor provided before grant (corrected)

Inventor name: KENNEDY, RICHARD

Inventor name: GOFFARD, NICOLAS

Inventor name: DAVISON, TIMOTHY

Inventor name: HILL, LAURA A.

Inventor name: HARKIN, DENIS PAUL

Inventor name: KEATING, KATHERINE E.

Inventor name: BYLESJO, MAX

Inventor name: DEHARO, STEVE

Inventor name: O'DONNELL, JUDE

Inventor name: PATTERSON, FIONNUALA

Inventor name: PROUTSKI, VITALI

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602011069113

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: G01N0033480000

Ipc: C12Q0001688600

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1186771

Country of ref document: HK

RIN1 Information on inventor provided before grant (corrected)

Inventor name: HILL, LAURA A.

Inventor name: KENNEDY, RICHARD

Inventor name: O'DONNELL, JUDE

Inventor name: GOFFARD, NICOLAS

Inventor name: HARKIN, DENIS PAUL

Inventor name: PROUTSKI, VITALI

Inventor name: PATTERSON, FIONNUALA

Inventor name: BYLESJO, MAX

Inventor name: KEATING, KATHERINE E.

Inventor name: DAVISON, TIMOTHY

Inventor name: DEHARO, STEVE

INTC Intention to grant announced (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: C12Q 1/6886 20180101AFI20200416BHEP

INTG Intention to grant announced

Effective date: 20200429

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602011069113

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1328283

Country of ref document: AT

Kind code of ref document: T

Effective date: 20201115

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1328283

Country of ref document: AT

Kind code of ref document: T

Effective date: 20201028

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20201028

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210128

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210301

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210129

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210128

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210228

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602011069113

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20210729

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20210930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210228

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210915

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210915

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210930

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210930

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20110915

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230516

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230918

Year of fee payment: 13

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230915

Year of fee payment: 13

Ref country code: DE

Payment date: 20230921

Year of fee payment: 13

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201028